atherosclerosis in progeria

117
ATHEROSCLEROSIS IN PROGERIA: INSIGHT FROM NEW MOUSE MODELS WITH SYSTEMIC AND TISSUE-SPECIFIC PROGERIN EXPRESSION MAGDA RITA HAMCZYK Madrid, 2017 UNIVERSIDAD AUTÓNOMA DE MADRID Departamento de Bioquímica

Upload: others

Post on 28-Apr-2022

9 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: ATHEROSCLEROSIS IN PROGERIA

ATHEROSCLEROSIS IN PROGERIA:

INSIGHT FROM NEW MOUSE MODELS WITH SYSTEMIC AND

TISSUE-SPECIFIC PROGERIN EXPRESSION

MAGDA RITA HAMCZYK

Madrid, 2017

UNIVERSIDAD AUTÓNOMA DE MADRID

Departamento de Bioquímica

Page 2: ATHEROSCLEROSIS IN PROGERIA

Departamento de Bioquímica

Facultad de Medicina

Universidad Autónoma de Madrid

Atherosclerosis in progeria: insight from new mouse models with systemic and tissue-specific

progerin expression

PhD student: Magda Rita Hamczyk, M.Sc. in Biotechnology

Thesis directors: Vicente Andrés, PhD and Ricardo Villa-Bellosta, PhD

Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)

Page 3: ATHEROSCLEROSIS IN PROGERIA

D. Vicente Andrés García, Doctor en Biología, y D. Ricardo Villa Bellosta, Doctor por la

Universidad de Zaragoza,

CERTIFICAN:

Que Dña. Magda Rita Hamczyk, licenciada en Biotecnología por la Universidad Jaguelónica

(Polonia) ha realizado bajo su dirección el trabajo de Tesis Doctoral que lleva por título

“Atherosclerosis in progeria: insight from new mouse models with systemic and tissue-specific

progerin expression”.

Revisado el presente trabajo, expresan su conformidad para la presentación del mismo por considerar

que reúne los requisitos necesarios para ser sometido a discusión ante el Tribunal correspondiente, para

optar al grado de Doctora.

En Madrid, a 26 de Junio de 2017

Dr. Vicente Andrés García Dr. Ricardo Villa Bellosta

Page 4: ATHEROSCLEROSIS IN PROGERIA

ACKNOWLEDGEMENTS

Page 5: ATHEROSCLEROSIS IN PROGERIA

This thesis would not have been possible without the help of many people. Thus, I would like to express

my sincere gratitude to:

Dr. Vicente Andrés, my thesis director, for giving me the opportunity to perform my PhD at

the CNIC and for all the support during more than 5 years of hard work and sacrifice (it would

be impossible to list all the tasks that were accomplished and all the obstacles that were

overcome during this period).

Dr. Ricardo Villa-Bellosta, my thesis co-director, labmate and friend, for believing in me

and being there for me 24/7. I am grateful to have been a part of this unstoppable

arteriosclerosis duo; thank you for the motivation, immense knowledge and for teaching me

the essential lab (survival) skills.

Dr. Carlos López-Otín for giving me the opportunity to develop a part of my project during

a (too) short stay at the University of Oviedo. Thank you for all the precious support in this

ER stress story. Without your help, it would not have been possible to conduct this research;

and thank you for giving me hope that a scientist does not have to sacrifice their personal life

to be successful.

Dr. Antonio Maraver and Dr. Andrés Hidalgo, my thesis committee members, for their

insightful comments and encouragement, and also for the hard questions which stimulated

me to improve my research. And thank you Andrés for cheering me up during my PhD blues

and always finding the time to talk to me even when you were up to your neck in work.

Dr. Krzysztof Guzik, my former research supervisor, for encouraging me to pursue a career

in research and providing advice during my postgraduate work.

María Jesús Andrés for the priceless technical support in many tedious laboratory

techniques (genotyping, lesion quantification, etc.) and for being like a mother to me.

Dr. Pilar Gonzalo for the optimism and invaluable help during the last year of my PhD which

allowed me to focus on writing my thesis and research articles; without you, it would not

have been possible to complete the project on time.

Antonio de Molina and Roisin Doohan for the support with histology. Thank you for sharing

your enthusiasm for exciting aortic and cardiac pathologies; I wish everybody had the same

commitment and love for their work as you have.

Rubén Mota for being the best vet for my mice and for teaching me many amazing things

about mouse pathology.

Page 6: ATHEROSCLEROSIS IN PROGERIA

The animal facility technicians, especially Virginia Zorita and Eva Santos, for the laborious,

day-to-day animal care.

Marta García Camacho and Ana Belén Ricote for the support with hematology, serum

biochemistry, ECG and blood pressure techniques.

Dr. Beatriz Dorado for keeping the lab in check and for the hard labor of managing the

group.

Eduardo Bieger and Eeva Soininen for the invaluable help with the Spanish bureaucracy.

Javier Mateos for the technical support.

My students, Lucia, Carmen, Miriam and Rosa, for teaching me how to be a leader rather

than a boss, for asking adequate questions, and for making me think out of the box.

Former (Carlos, Pedro, Laia, Raphael, Alba, Óscar, José) and current lab members (José

María, Raquel, Amanda, Víctor, Alberto, Lara, Álvaro, Elba, Cristina) for the

stimulating discussions and all the time spent together at the CNIC.

CNIC´s Polish team (Magda, Ola, Dori, and Ania) for sharing those good and bad moments

on this PhD emotional rollercoaster, for all the scientific and non-scientific support, pierogi

cooking, pączki eating and much more.

Carlos López-Otín´s group members for the warm reception at the University of Oviedo

during my internship.

Riju for entertaining me during our 12:45 lunch sessions. Thank you for making me believe

that there are still good people out there.

Víctor, my soul-mate, for being a true and great emotional-scientific supporter at all times,

and for telling awful jokes that made me forget how bad the situation was. Thank you for

being non-judgmental of me and instrumental in instilling confidence, and for inspiring me

to strive towards my goal.

Maria, my flatmate and friend, for all the time spent together talking, laughing, dancing,

cooking; thank you for becoming a part of my life.

Ania, Roman, Sofia and Maxim for being my eastern family here in Madrid; thank you for

sharing many precious moments with me.

My parents for their love and encouragement, for providing me with the best education they

could, and most importantly for teaching me how to think rather than what to think.

My siblings (Sylwia and Rafał) and friends back in Poland (Łukasz, Justyna, Gosia,

Monika, Madzia and many others) for putting up with me and my endless excuses related

Page 7: ATHEROSCLEROSIS IN PROGERIA

to continuous lack of time. Special thanks to Łukasz for encouraging me to relax and enjoy

life during the most difficult moments of my PhD and for being the best friend I could dream

of.

Page 8: ATHEROSCLEROSIS IN PROGERIA

RESUMEN

Page 9: ATHEROSCLEROSIS IN PROGERIA

Las enfermedades cardiovasculares (ECV) son la causa principal de morbimortalidad en el mundo

debido en gran parte al envejecimiento progresivo de nuestras sociedades. El deterioro cardiovascular

está acelerado en un trastorno genético muy raro llamado síndrome de progeria Hutchinson-Gilford

(HGPS). Esta enfermedad está causada por una mutación puntual de novo en el gen LMNA, que conduce

a la expresión de “progerina”, una forma mutante de la lamina A que provoca numerosas anomalías

estructurales y funcionales en el núcleo. Los niños con HGPS presentan síntomas de envejecimiento

prematuro, incluyendo alopecia, osteoporosis, lipodistrofia, rigidez de las articulaciones, arrugas de la

piel y moteado, siendo la característica más importante de la enfermedad la aterosclerosis acelerada,

proceso que conduce a muerte prematura a una edad media de 14,6 años, predominantemente por

infarto de miocardio o accidente cerebrovascular. Existe un gran desconocimiento sobre los

mecanismos por los que la progerina acelera la aterosclerosis, en gran parte debido a la falta de modelos

animales adecuados. En esta tesis hemos generado nuevos modelos de ratón que permiten estudiar la

aterosclerosis asociada a HGPS. Comparado con controles con el gen Lmna intacto, los ratones

mutantes que expresan progerina de forma ubicua presentan envejecimiento prematuro asociado con

pérdida de peso corporal y menor longevidad. Además, mostraron patología vascular similar a la

observada en pacientes HGPS, incluyendo aterosclerosis acelerada, pérdida de células de músculo liso

vascular (CMLVs), aumento del contenido de colágeno, retención de lípidos en la capa media y fibrosis

de la capa adventicia. También hemos demostrado que los ratones que expresan progerina

específicamente en CMLVs, pero no en macrófagos, recapitulan la patología vascular del modelo

ubicuo. Además, tanto en el modelo ubicuo como en el específico de CMLVs, los ateromas mostraron

evidencia de ruptura de la placa que podría provocar infarto de miocardio. Por otra parte, mediante

transcriptómica de alto rendimiento identificamos en ambos modelos de ratón el estrés de retículo

endoplásmico (RE) y la respuesta a proteínas desplegadas como posibles mecanismos inductores de

muerte de CMLVs y aterosclerosis acelerada. De acuerdo con esta hipótesis, el tratamiento con ácido

tauroursodeoxicólico (TUDCA), una chaperona química que aumenta la capacidad celular para

soportar el estrés de RE, disminuyó en ambos modelos la aterosclerosis, la pérdida de CMLVs y el

grosor de la adventicia. TUDCA también prolongó en un 35% la supervivencia de los ratones con

expresión de progerina específica en CMLVs. En resumen, nuestros resultados sugieren el uso de

TUDCA para prevenir la aterosclerosis y eventos cardiovasculares asociados a HGPS. Dado que la

progerina se acumula con la edad en individuos sin HGPS, nuestros resultados también podrían arrojar

luz sobre el envejecimiento fisiológico.

Page 10: ATHEROSCLEROSIS IN PROGERIA

SUMMARY

Page 11: ATHEROSCLEROSIS IN PROGERIA

Cardiovascular disease (CVD) is a major cause of morbidity and mortality worldwide due to the

progressive aging of our societies. Age-related decline in cardiovascular health is accelerated in a rare

genetic disorder called Hutchinson-Gilford progeria syndrome (HGPS). The disease is caused by a de

novo point mutation in the LMNA gene, which leads to the expression of “progerin”, a mutant form of

the nuclear protein lamin A. Since lamin A possesses important structural and functional properties,

progerin expression triggers numerous nuclear abnormalities. Children with HGPS exhibit premature

aging symptoms, including alopecia, osteoporosis, lipodystrophy, joint stiffness, and skin wrinkling

and mottling. However, the most clinically relevant feature of the disease is accelerated atherosclerosis,

which leads to premature death at an average age of 14.6 years, predominantly from myocardial

infarction or stroke. The mechanisms through which progerin provokes enhanced atherosclerosis

remain poorly understood, in part due to the paucity of suitable models. To address this, we sought to

generate new mouse models that allow the study of atherosclerosis in the context of HGPS. When

compared with control mice expressing wild-type lamin A/C, mice with ubiquitous progerin expression

exhibited a premature aging phenotype, including reduced body weight and shortened survival. In

addition, progerin-expressing mice showed increased atherosclerosis burden together with a severe

vascular pathology, including the depletion of vascular smooth muscle cells (VSMCs), increased

collagen content, medial lipid retention and adventitial fibrosis, resembling most aspects of CVD

observed in HGPS. We also found that mice expressing progerin specifically in VSMCs, but not in

macrophages, fully recapitulated the vascular pathology observed in the ubiquitous progeria model.

Atheromas of both ubiquitous and VSMC-specific models showed evidence of plaque disruption,

which might lead to myocardial infarction. Using a transcriptomic approach, we identified endoplasmic

reticulum (ER) stress and the unfolded protein response as possible driver mechanisms of progerin-

induced VSMC death and accelerated atherosclerosis. Accordingly, treatment with

tauroursodeoxycholic acid (TUDCA), a chemical chaperone that increases the capacity of a cell to

sustain ER stress, was effective at ameliorating vascular disease (atherosclerosis, VSMC loss and

adventitial thickening) in both ubiquitous and VSMC-specific mouse models. TUDCA also prolonged

the survival of mice with VSMC-specific progerin expression by 35%. Taken together, these findings

indicate that TUDCA may be effective in the treatment of atherosclerosis and associated cardiovascular

events in HGPS. Moreover, since progerin accumulates with age in non-HPGS individuals, our data

may also shed some light on the mechanisms of normal aging.

Page 12: ATHEROSCLEROSIS IN PROGERIA

INDEX

Page 13: ATHEROSCLEROSIS IN PROGERIA

ABBREVIATIONS .............................................................................................................................. 1

I. INTRODUCTION ............................................................................................................................ 7

I.1. Physiological aging and cardiovascular disease (CVD) .......................................................... 9

I.1.1. Aging as a risk factor for CVD ........................................................................................... 9

I.1.2. Atherosclerosis ..................................................................................................................... 9

I.2. Premature aging and CVD ...................................................................................................... 10

I.2.1. Hutchinson-Gilford progeria syndrome (HGPS) ........................................................... 10

I.2.2. CVD in HGPS .................................................................................................................... 11

I.2.3. Mechanism leading to progerin production .................................................................... 13

I.3. Progerin and prelamin A in physiological aging ................................................................... 15

I.4. Mouse models of progeria ....................................................................................................... 15

I.5. General mechanisms of HGPS ................................................................................................ 17

I.6. Mechanisms underlying CVD in progeria ............................................................................. 19

I.6.1. VSMC loss .......................................................................................................................... 19

I.6.2. Vascular calcification ........................................................................................................ 21

I.6.3. Endothelial dysfunction .................................................................................................... 22

I.6.4. Cardiac electrical alterations............................................................................................ 22

I.7. Endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) ................. 23

II. OBJECTIVES................................................................................................................................ 27

III. MATERIALS AND METHODS ................................................................................................ 31

Page 14: ATHEROSCLEROSIS IN PROGERIA

III.1. Mice ......................................................................................................................................... 33

III.2. Longevity study ...................................................................................................................... 33

III.3. High-fat diet (HFD) experiments ......................................................................................... 33

III.4. Hematology and serum biochemical analysis ..................................................................... 34

III.5. Blood pressure measurement ............................................................................................... 34

III.6. ECG ........................................................................................................................................ 34

III.7. Treatment ............................................................................................................................... 35

III.8. Quantification of atherosclerosis burden ............................................................................ 35

III.9. Histology and immunofluorescence ..................................................................................... 35

III.10. Sample collection and preparation for RNAseq ............................................................... 36

III.11. RNAseq library preparation, sequencing, and generation of FastQ files ...................... 37

III.12. Differential expression analysis .......................................................................................... 37

III.13. Pathway analysis .................................................................................................................. 38

III.14. RNA extraction and cDNA preparation ............................................................................ 38

III.15. PCR detection of lamin A and progerin ............................................................................ 38

III.16. Quantitative real-time PCR ................................................................................................ 39

III.17. Statistical analysis ................................................................................................................ 39

IV. RESULTS AND DISCUSSION ................................................................................................... 41

IV.1. Ubiquitous progerin expression aggravates atherosclerosis in HFD-fed Apoe-/- mice .... 43

IV.2. VSMC-specific progerin expression in HFD-fed Apoe-/- mice aggravates atherosclerosis

and shortens lifespan....................................................................................................................... 48

Page 15: ATHEROSCLEROSIS IN PROGERIA

IV.3. Progerin triggers plaque vulnerability ................................................................................ 57

IV.4. Progerin expression accelerates atherosclerosis in Apoe-/- mice fed normal chow .......... 59

IV.5. Progerin expression in VSMCs leads to progressive hypotension .................................... 62

IV.6. Apoe-/-LmnaG609G/G609G mice develop arrhythmias .............................................................. 64

IV.7. Apoe-/-LmnaLCS/LCSSM22Cre die from atherosclerosis-related causes ............................ 66

IV.8. Progerin expression in VSMCs activates endoplasmic ER stress and the UPR .............. 72

IV.9. Therapeutic effects of ER stress response targeting in progeroid mice ........................... 78

V. CONCLUSIONS ............................................................................................................................ 87

V. CONCLUSIONES ......................................................................................................................... 93

VI. REFERENCES ............................................................................................................................. 99

VII. ANNEX ...................................................................................................................................... 121

Page 16: ATHEROSCLEROSIS IN PROGERIA

1

ABBREVIATIONS

Page 17: ATHEROSCLEROSIS IN PROGERIA

ABBREVIATIONS

3

53BP1 - 53 binding protein-1

APOE - apolipoprotein E

ATF4 - activating transcription factor 4

ATF6 - activating transcription factor 6

ATF6f - ATF6 fragment

BAC - bacterial artificial chromosome

BMP2 - bone morphogenetic protein 2

BSA - bovine serum albumin

CVD - cardiovascular disease

DDIT3 - DNA damage-inducible transcript 3

DNA-PK - DNA-dependent protein kinase

DNA-PKcs - DNA-dependent protein kinase catalytic subunit

EC(s) - endothelial cell(s)

ECG - electrocardiogram

eIF2α - eukaryotic translation initiator factor 2α

ePPi - extracellular inorganic pyrophosphate

ER - endoplasmic reticulum

ERAD - ER-associated protein degradation

FTI(s) - farnesyl transferase inhibitor(s)

GRP78 - 78 kDa glucose-regulated protein

H&E - hematoxylin-eosin

HDL - high-density lipoproteins

HFD - high-fat diet

HGPS - Hutchinson-Gilford progeria syndrome

Page 18: ATHEROSCLEROSIS IN PROGERIA

ABBREVIATIONS

4

ICMT - isoprenylcysteine carboxyl methyltransferase

IPA - Ingenuity Pathway Analysis

iPSC(s) - induced pluripotent stem cell(s)

IRE1 - inositol-requiring enzyme 1

LDL - low-density lipoproteins

mTOR - mammalian target of rapamycin

NHEJ - non-homologous end joining

ORO - Oil Red O

PBA - 4-phenyl butyric acid

PBS - phosphate-buffered saline

PERK - protein kinase RNA-like ER kinase

RT - room temperature

RUNX2 - Run-related transcription factor-2

SEM - standard error of the mean

SMC(s) - smooth muscle cell(s)

TUDCA - tauroursodeoxycholic acid

UPR - unfolded protein response

VSMC(s) - vascular smooth muscle cell(s)

XBP1 - X box-binding protein 1

XBP1s - spliced X box-binding protein 1

Page 19: ATHEROSCLEROSIS IN PROGERIA

7

I. INTRODUCTION

Page 20: ATHEROSCLEROSIS IN PROGERIA

I. INTRODUCTION

9

I.1. Physiological aging and cardiovascular disease (CVD)

I.1.1. Aging as a risk factor for CVD

Aging is the main risk factor for the development of CVD (1). Although much progress has

been made in the prevention, diagnosis, and treatment of CVD, it remains the leading cause of death

worldwide (2). According to the World Health Organization, approximately 17.5 million people die

annually from CVD, representing 31% of all deaths (www.who.int/cardiovascular_diseases). CVD is

the outcome of complex interactions between modifiable and non-modifiable risk factors (3). Many

modifiable risk factors for CVD have been identified, and the causal relevance of several of these

factors is now well proven, including tobacco exposure, physical inactivity, obesity, hypertension,

hypercholesterolemia and diabetes. Non-modifiable risk factors include age, gender, ethnicity and

genetic susceptibility to the disease (family history).

Age-related changes in the vasculature include luminal dilatation and arterial wall thickening,

especially of the intima (e.g. atherosclerosis), leading to increased vascular stiffness (1, 4). Various

changes in the vessel wall that show causal relationships with arterial stiffening have been described,

including calcification, augmented content and crosslinking of collagen fibers, increased elastin

breakage, and diminished elastin content (5-8). Moreover, endothelial function is impaired with age,

further contributing to the reduced vascular compliance (1), defined as the capacity of a blood vessel

to enlarge and contract in response to changes in the pressure.

I.1.2. Atherosclerosis

Atherosclerosis underlies most manifestations of CVD, including coronary heart disease and

cerebrovascular disease, and can lead to myocardial infarction or stroke. Atherosclerosis is

characterized by thickening of the arterial wall and luminal narrowing due to atherosclerotic plaque

buildup. The disease is initiated by endothelial dysfunction triggering low-density lipoprotein (LDL)

infiltration and accumulation in the intima, principally at sites with disturbed blood flow (9). The

presence of proteoglycans in the subendothelial space can further increase the retention of LDL

particles (10), which are subsequently oxidized by reactive oxygen species. Oxidized LDL induces

endothelial activation, which is characterized by the expression of adhesion molecules and the secretion

of chemokines that trigger the recruitment of monocytes to the intima and their differentiation into

macrophages. Neointimal macrophages begin to ingest modified LDL via the scavenger receptor

Page 21: ATHEROSCLEROSIS IN PROGERIA

I. INTRODUCTION

10

pathway (11), leading to their transformation into foam cells that secrete pro-inflammatory molecules,

recruiting more immune cells to the lesion site. Impairment in the processing of oxidized lipoproteins

leads to foam cell death (12). Progressive accumulation of cell debris and cholesterol crystals results

in the formation of a necrotic core. Growth factors and cytokines present within the lesion induce the

phenotypic transition of vascular smooth muscle cells (VSMCs) from a “contractile” to a “synthetic”

state, and their migration from the media to the growing neointimal lesion. Neointimal synthetic

VSMCs proliferate and abundantly synthesize extracellular matrix, normally stabilizing plaques.

Mature fibroatheromatous plaques have a collagen- and VSMC-rich fibrous cap covering the necrotic

core, which prevents its rupture and the subsequent life-threatening thrombus formation.

Plaque destabilization, including plaque rupture, plaque erosion and the formation of calcified

nodules, is a complex process that depends on both structural features and biomechanical forces. Plaque

rupture, which is the best characterized and the most common manifestation of plaque destabilization,

is characterized by fibrous cap disruption and the ensuing exposure of the necrotic core to blood

constituents, resulting in thrombus formation. Plaque destabilization may also occur due to plaque

erosion and the accumulation of calcified nodules. Atherosclerotic lesions that are prone to rupture are

termed “vulnerable” plaques, and are usually characterized by large necrotic cores, thin fibrous caps

(related to decreased collagen and VSMC content), intraplaque hemorrhage, neoangiogenesis and large

inflammatory infiltrates (13). Since plaque disruption can lead to myocardial infarction or stroke, it is

of considerable importance to characterize and detect vulnerable plaques in order to prevent

cardiovascular events in patients.

I.2. Premature aging and CVD

I.2.1. Hutchinson-Gilford progeria syndrome (HGPS)

HGPS is an extremely rare disorder characterized by signs of premature aging. This devastating

disease was first described by Jonathan Hutchinson in 1886 and Hastings Gilford in 1897 (14, 15). The

prevalence of HGPS is 1 in 20 million and it has no ethnic or gender bias (16). Affected children are

normal at birth but within the first two years of life they begin to present the first symptoms of the

disease, such as failure to thrive and skin abnormalities. Patients progressively develop other signs of

premature aging, including alopecia, mottled and wrinkled skin, loss of body fat, joint stiffness and

osteoporosis (16, 17). Importantly, they show generalized atherosclerosis, which leads to death from

Page 22: ATHEROSCLEROSIS IN PROGERIA

I. INTRODUCTION

11

myocardial infarction or stroke at an average age of 14.6 years (18). While HGPS resembles most

aspects of normal aging, patients do not show signs of neurodegenerative disease or cancer.

I.2.2. CVD in HGPS

The most clinically relevant feature of HGPS is atherosclerosis, which is responsible for more

than 90% of deaths (16). Atherosclerosis in HGPS is particularly intriguing because patients lack the

classical risk factors for CVD development, such as hypercholesterolemia and increased serum C-

reactive protein (19). Although levels of athero-protective high-density lipoproteins (HDL) are similar

between prematurely-aged and healthy children, there is an age-dependent decline in HDL levels in

HGPS (19). Autopsy findings have revealed that patients with HGPS present a wide spectrum of plaque

phenotypes, ranging from early to late stage (20). These atherosclerotic lesions usually exhibit

calcification, inflammation, and evidence of plaque erosion or rupture, similar to what is observed in

physiological aging (20). The overall absence of pronounced dyslipidemia in progeria could partially

explain why HGPS lesions tend to have smaller atheromatous cores than those of typical age-associated

atheromas (20).

The severity of atherosclerosis in progeria usually correlates with VSMC depletion in the media

(21, 22). This severe loss of VSMCs is accompanied by extracellular matrix deposition and altered

elastin structure (20-22). In contrast to what is seen in normal aging, HGPS arteries and veins present

prominent thickening of the adventitia (20), which together with VSMC loss may result in vessel

stiffening and reduced vascular compliance, triggering plaque development. Accordingly, patients with

HGPS exhibit an increase in both carotid-femoral pulse wave velocity and arterial wall echodensity,

confirming that vascular stiffening is an essential contributor to cardiovascular decline in progeria (23).

Aortic stiffness can then lead to augmented afterload and the subsequent left ventricular hypertrophy

observed in some HGPS patients (16, 17, 20, 22).

The ability of blood vessels to stretch in response to changes in the pressure plays an important

role in the regulation of blood pressure. Although some authors reported development of hypertension

in HGPS subjects, it does not seem to be a generalized symptom of the disease. Merideth et al. described

elevated systolic or diastolic blood pressures in 7/15 patients as compared with both age- and height-

matched healthy controls (17). However, they reported only the highest reading even though blood

pressure was measured an average of six times during the course of the study, which questions the

validity of their results and conclusions. Further work by Gerhard-Herman et al. reported increased

Page 23: ATHEROSCLEROSIS IN PROGERIA

I. INTRODUCTION

12

systolic pressure in 7/26 patients and increased diastolic pressure in 9/26 patients as compared with

age-matched controls (23). When they applied height-age standards to account for the growth

impairment, systolic and diastolic blood pressures were higher in approximately half of the patients.

Nonetheless, pulse pressure, which is defined as the difference between systolic and diastolic blood

pressure and positively correlates with CVD risk during physiological aging (24, 25), was within the

normal range for both chronologic age and height age (23). Of note, none of the studies analyzed

hypotension incidence in HGPS, albeit raw data from some reports clearly show that some blood

pressure measurements are below the normal range for the age-matched controls (20).

These somewhat contradictory observations on blood pressure may in fact indicate that progeria

patients have blood pressure instability and variability, which has been recently shown to be a strong

predictor of stroke (26, 27). This could be of special relevance for HGPS because arterial ischemic

strokes are common during the disease progression and may lead to death in about 10% of patients

(18). A retrospective study by Silvera et al. found evidence of stroke in 60% of patients, of which half

was clinically silent (28). Likewise, vascular alterations in the brain, including intracranial and distal

vertebral artery steno-occlusive disease with collateral vessel formation, are frequently found in HGPS

(28). Remarkably, this arteriopathy is different from other vasculopathies of childhood and

cerebrovascular disease during physiological aging (28). In spite of evident vascular alterations in the

brain, progeric patients have normal cognitive function with the exception of stroke-related defects (16,

29).

While stroke accounts for some mortality in HGPS, the most common cause of death is

myocardial infarction. As in the case of stroke, patients may have clinically silent myocardial infarcts

during their lifetime (16). Indeed, autopsy findings provided evidence of remote (months), subacute (2-

3 weeks), and acute (2-3-day-old) myocardial infarctions in the same patient (20). These acute and

chronic infarcts may lead to arrhythmia and death. In a recent study, Rivera-Torres et al. showed age-

related repolarization abnormalities in HGPS patients, manifested as ST segment depression/elevation

and negative or biphasic T waves on the electrocardiogram (ECG), which can lead to an increased risk

of arrhythmias (30). Another study also reported age-related ECG abnormalities in some patients, such

as prolonged QT interval (17). Cardiac electrical defects in HGPS may be a consequence of heart

fibrosis caused by non-fatal infarcts as well as valvular dysfunction. Likewise, various studies

described severe degenerative aortic and mitral valvular disease in HGPS, characterized by thickening,

fibrosis and calcification of the valve leaflets (16, 17, 20, 31, 32).

Page 24: ATHEROSCLEROSIS IN PROGERIA

I. INTRODUCTION

13

In summary, HGPS patients present severe cardiovascular defects in the vessels, heart, brain

and valves, which resemble many aspects of physiological aging. Thus, progeria is a model of

cardiovascular aging isolated from classical cardiovascular risk factors and may enhance our

knowledge about the initiation and progression of vascular stiffening, atherosclerosis and the incidence

of cardiovascular events.

I.2.3. Mechanism leading to progerin production

HGPS is caused by a heterozygous de novo point mutation within the LMNA gene (33, 34). In

normal cells, alternative splicing of the LMNA gene gives rise to two major isoforms, lamin A and

lamin C (35). Some minor forms are also produced, such as lamin A10 and the germline-specific

lamin C2 variant (36, 37). Lamin A and lamin C are expressed in the majority of differentiated somatic

cells, and play important roles in maintaining nuclear structure and function (38).

Various posttranslational modifications of prelamin A (664 amino acid residue) give rise to

mature lamin A (646 amino acid residue) (Fig. 1). First, the CaaX motif located at the C-terminus of

prelamin A is farnesylated by farnesyl protein transferase (39, 40). Next, the last three residues (-aaX)

are removed usually by the zinc metallopeptidase STE24 (ZMPSTE24), also known as farnesylated

proteins-converting enzyme 1 (40). The newly exposed C-terminal cysteine is then carboxymethylated

by isoprenylcysteine carboxylmethyltransferase (ICMT) (40). These modifications are believed to

enhance hydrophobic interactions with the inner nuclear membrane and to facilitate assembly into the

nuclear lamina (41). Finally, ZMPSTE24 cleaves the last 15 amino acids together with the farnesyl and

carboxymethyl groups (after tyrosine in position 646), resulting in mature lamin A (42-44).

Page 25: ATHEROSCLEROSIS IN PROGERIA

I. INTRODUCTION

14

Figure 1. Lamin A maturation. Lamin A is formed from a precursor termed prelamin A. Prelamin A is first farnesylated

by a farnesyltransferase (FT) on the cysteine residue within the CaaX motif (where “C” is cysteine, “a” is an aliphatic amino

acid, and “X” is variable, in case of prelamin A: CSIM). Next, the C-terminal tripeptide (–SIM) is removed, usually by

ZMPSTE24, a zinc metalloendoprotease. This is followed by carboxymethylation of the newly-exposed cysteine by

isoprenylcysteine carboxylmethyltransferase (ICMT). Finally, the last 15 amino acids containing the farnesyl and

carboxymethyl modifications are cleaved by ZMPSTE24, resulting in mature lamin A.

“Classical” HGPS is caused by a single nucleotide substitution in exon 11 (c.1824C>T;

p.G608G) (33, 34). This cytosine-to-thymine substitution does not change the amino acid encoded

(both GGC and GGT codons encode glycine), but activates a cryptic splice site leading to a deletion of

150 nucleotides in the mRNA. This aberrant splicing results in the production of a truncated form of

lamin A, referred to as progerin, missing 50 amino acids close to the C-terminus. Progerin lacks the

cleavage site for ZMPSTE24 and therefore retains the last 15 residues together with farnesyl and

carboxymethyl groups. Permanently farnesylated and carboxymethylated progerin stays anchored in

the inner nuclear membrane, leading to abnormalities in the nuclear shape and function (45, 46).

Page 26: ATHEROSCLEROSIS IN PROGERIA

I. INTRODUCTION

15

I.3. Progerin and prelamin A in physiological aging

Under normal conditions, prelamin A is almost undetectable in cells because it is rapidly

processed into mature lamin A after translation. Farnesylated prelamin A accumulation due to

inactivating mutations in the ZMPSTE24 gene also leads to progeria-like syndromes, such as restrictive

dermopathy (47-49) and mandibuloacral dysplasia (50). Remarkably, increased amounts of prelamin

A have been also observed during physiological aging, presumably due to an age-dependent

downregulation of ZMPSTE24 (51). Interestingly, inhibitors of proteases used for human

immunodeficiency virus treatment result in increased levels of prelamin A and accelerated vascular

aging (52, 53).

Many studies revealed that non-HGPS individuals accumulate small amounts of progerin with

age (20, 54, 55). One possible explanation for this is because in healthy subjects the cryptic donor splice

site shares 5 of 7 bases with the consensus splice sequence, leading to sporadic usage of the splice site.

In HGPS, the cryptic donor splice site shares 6 of 7 nucleotides with the consensus splice sequence due

to the C-to-T substitution, resulting in the frequent occurrence of aberrant splicing.

Thus, the accumulation of permanently farnesylated prelamin A and progerin in both normal

and premature aging strongly suggests that some mechanisms are shared between these physiological

and pathological processes. Accordingly, conclusions derived from the study of progeria may also shed

some light on normal aging.

I.4. Mouse models of progeria

As of 1st April 2017, there were 148 identified children living with progeria worldwide

(www.progeriaresearch.org). Of these, 115 carry the HGPS classical mutation, and the remaining 33

children have a mutation within the lamin A pathway but do not express progerin. This extremely low

number of HGPS patients leads to challenges in conducting both clinical trials and research on the

mechanisms driving the disease. Because of this, various mouse models of progeria have been created

over the past 15 years that resemble many, but not all, premature aging features. This may be in part

attributed to interspecies differences (e.g., in lipid metabolism) and distinct toxicity of progerin in mice

and humans. Furthermore, the differences in the phenotype between different mouse models may be a

consequence of diverse strategies used during their generation, which lead to variable progerin levels,

as well as the absence or presence of other Lmna gene products, such as lamin A and lamin C.

Page 27: ATHEROSCLEROSIS IN PROGERIA

I. INTRODUCTION

16

The first progerin-expressing mouse model was developed by Yang et al. in 2005. The authors

created a mutant allele called LmnaHG (“Hutchinson-Gilford”) that, unlike in the human disease, gives

rise exclusively to progerin (56, 57). Heterozygous LmnaHG/+ mice, which express progerin from the

mutant allele and lamin A/C from the wild-type allele, showed reduced weight gain around 6-8 weeks

of age and died by 4-6 months of age (compared with a lifespan of more than 2 years for wild-type

mice). Homozygous LmnaHG/HG mice expressing solely progerin presented a more severe aging

phenotype and died by 3-4 weeks of age. Both, LmnaHG/+ and LmnaHG/HG mice displayed some of the

symptoms common to HGPS, such as hair loss, osteoporosis, loss of subcutaneous fat, but no

cardiovascular alterations were observed (57).

Varga et al. generated a transgenic mouse model by introducing a bacterial artificial

chromosome (BAC) containing the human LMNA gene carrying an HGPS-causing mutation

(c.1824C>T; p.G608G) (58). In addition to the mutated LMNA gene, the BAC also contained RAB25

(coding for another isoprenylated protein), UBQLN4 and MAPBPIP genes. In consequence, G608G

BAC mice expressed mouse endogenous lamin A/C together with human progerin and 3 other human

proteins. While these mice did not present any evident signs of premature aging, progressive VSMC

depletion was observed in the media, beginning at 5 months of age. Reduced VSMC number was

accompanied by an increased deposition of collagen and proteoglycan in the media, altered elastic fiber

structure, and thickened adventitial and medial layers. Remarkably, the vascular pathology was similar

to that observed in HGPS (20-22). In line with the aortic phenotype, vascular responsiveness to sodium

nitroprusside administration was decreased in G608G BAC mice. Nevertheless, no evidence of

atherosclerotic plaque formation was found (58).

LmnaHG and G608G BAC mice fail to fully recapitulate the human disease, showing only

premature aging or vascular alterations. Thus, Osorio et al. created a knock-in mouse model harboring

a c.1827C>T (p.G609G) mutation within the mouse Lmna gene, which is equivalent to the human

c.1824C>T (p.G608G) HGPS-causing single nucleotide substitution (59). Similar to what is observed

in HGPS, the presence of the LmnaG609G allele leads to the synthesis of progerin via aberrant splicing,

together with lamin C and some residual lamin A. Homozygous LmnaG609G/G609G mice showed growth

retardation after 3 weeks of age and died at an average age of 15 weeks. They also presented a loss of

subcutaneous fat, attrition of hair follicles, and bone alterations. Notably, LmnaG609G/G609G mice

presented VSMC depletion in the medial layer of the aortic arch, but not of the thoracic aorta. Despite

the loss of VSMCs, no changes in the blood pressure were detected; however, animals developed age-

dependent bradycardia. Moreover, prolonged QRS duration was observed on the ECG, indicating

Page 28: ATHEROSCLEROSIS IN PROGERIA

I. INTRODUCTION

17

alterations in cardiac conduction. Heterozygous LmnaG609G/+ mice appeared normal until 8 months of

age, when they started presenting an aging-phenotype similar to that of homozygotes, and died at an

average age of 35 weeks. Remarkably, LmnaG609G/+ mice developed severe calcification of the aortic

media (60), a relevant symptom observed in HGPS. In summary, whereas LmnaG609G/G609G mice

recapitulate most of the clinical features of progeria, atherosclerosis has not been reported.

Lee et al. have recently created a new LmnaG609G/G609G mouse model that, similar to the model

developed by Osorio et al., harbors an HGPS-causing mutation in the Lmna gene that yields progerin

via abnormal splicing (61). Homozygous LmnaG609G/G609G mice display a phenotype similar to that of

other formerly described progeria models (43, 57, 59, 62), including VSMC depletion in the media of

the ascending aorta and adventitial fibrosis by 4 month of age. No detailed examination of the other

phenotypical characteristics was provided by the authors (61).

Other important models of premature aging are Zmpste24-deficient mice, which were generated

in parallel by two different groups (43, 62). These mice accumulate permanently-farnesylated prelamin

A due to a loss of function of the metalloproteinase involved in lamin A maturation. The Zmpste24-/-

mouse model created by Bergo et al. showed growth impairment, decreased subcutaneous fat content,

alopecia, muscle weakness and bone abnormalities, and died at 6-7 months of age (62). Zmpste24 null

mice generated by Pendas et al. presented a slightly more severe phenotype, characterized by growth

delay, lipodystrophy, alopecia, skeletal and muscular atrophy, cardiac abnormalities (heart interstitial

fibrosis, thinning of the ventricular wall and dilatation of both ventricles), and death at an average age

of 5 months (43). Consistent with the cardiac phenotype described by Pendas et al. (43), recent studies

revealed electrical cardiac alterations in Zmpste24-deficient mice (30).

I.5. General mechanisms of HGPS

Lamin A plays important roles in a wide variety of cellular functions, ranging from maintaining

the mechanical stability of the nucleus to regulating gene transcription and signal transduction (38).

Accordingly, accumulation of progerin and prelamin A may trigger the disease through various non-

mutually exclusive mechanisms. These mechanisms might differ across tissues due to the variable

amount of lamin A (and progerin) produced, depending on tissue stiffness (63, 64).

Progerin elicits its detrimental effects in a dose-dependent manner. Different progerin-causing

LMNA point mutations lead to a wide spectrum of disease severity, ranging from neonatal to late-onset

Page 29: ATHEROSCLEROSIS IN PROGERIA

I. INTRODUCTION

18

premature aging, which correlates with the expression level of progerin (65, 66). Thus, antisense

oligonucleotide treatment, which reduced the amount of progerin, ameliorated the vascular or aging

phenotype in two different LmnaG609G/G609G mouse models (59, 61).

The farnesyl group, which anchors progerin in the inner nuclear membrane, is believed to

account for at least part of its toxic effect. This hypothesis was initially supported by findings on

ZMPSTE24 deficiency in humans and mice, which results in the accumulation of farnesylated prelamin

A and causes various HGPS-like phenotypes (43, 47, 48, 62). The progeroid phenotype is milder in

patients that carry both a heterozygous LMNA mutation and a homozygous loss-of-function ZMPSTE24

mutation, possibly due to a reduced amount of farnesylated prelamin A (67). Likewise, Zmpste24-/-

mice with Lmna haplodeficiency do not present any evident aging phenotype (68). The importance of

farnesylation in the pathogenesis of HGPS was corroborated by the generation of LmnacsmHG/csmHG and

LmnanPLAO/nPLAO mouse models expressing nonfarnesylated progerin and prelamin A, respectively,

which did not exhibit a progeroid phenotype (69, 70). Furthermore, treatment with farnesyl transferase

inhibitors (FTIs) reverted the nuclear defects in progerin-expressing cells (56, 71), and prolonged the

lifespan of Zmpste24-/-and LmnaHG/+ mice (57, 72). FTIs also prevented CVD in progeroid G608G

BAC transgenic mice (73).

It was subsequently demonstrated that prelamin A and progerin can undergo alternative

prenylation by geranylgeranyltransferase upon farnesyltransferase inhibition (74). Thus, inhibition of

both farnesylation and geranylgeranylation with a combination of statins and aminobisphosphonates

was found to ameliorate the progeriod phenotype and extend survival in Zmpste24-/- mice (74). Based

on all the above findings, a clinical trial with HGPS patients was conducted targeting farnesylation with

an FTI (lonafarnib) alone or in combination with a statin (pravastatin) and a bisphosphonate

(zoledronate) (75-77). Monotherapy with lonafarnib improved vascular stiffness, bone structure, and

audiological status in some patients, and was estimated to extend survival by 1.6 years (75, 76). Triple-

drug therapy with lonafarnib, pravastatin and zoledronate had an additional beneficial effect on bone

mineral density, but no cardiovascular improvement was detected when compared with lonafarnib

monotherapy (77).

Prenylation (farnesylation and geranylgeranylation) is not the only detrimental effect of

progerin, since the aforementioned combination treatment provided some therapeutic benefits but not

a cure for HGPS. Supported by the observation that unfarnesylated progerin forms aggregates at the

nuclear membrane, akin to the farnesylated form, Kalinowski et al. recently suggested that increased

Page 30: ATHEROSCLEROSIS IN PROGERIA

I. INTRODUCTION

19

electrostatic interactions and aggregation are also responsible for progerin association with the nuclear

inner membrane (78). In addition, Qin et al. showed that progerin has a less heterogeneous and a more

compact tail compared with normal lamin A, and this may affect its interaction with DNA and other

proteins (79). Accumulating evidence shows that progerin may also elicit toxic effects through altered

protein structure due to the deletion of 50 amino acids at the C terminus. Abnormal interactions of

progerin with other nuclear components result in changes in nuclear shape (so-called nuclear blebbing),

increased thickness and stiffness of the lamina, heterochromatin mislocalization, and alterations in

nuclear pore complexes (80, 81). Lee et al. showed that progerin exhibits strong binding affinity for

lamin A/C and this interaction induced nuclear abnormalities (82). Importantly, pharmacological

therapy to disrupt the progerin-lamin A/C binding reduced nuclear aberrations and prevented cell

senescence in vitro, and also ameliorated progeroid features and extended lifespan in LmnaG609G/G609G

mice (82).

A recent study by Ibrahim et al. revealed that methylation of progerin by ICMT might also play

a role in premature aging syndromes (83). They showed that hypomorphic Zmpste24-/-Icmthm/hm mice

with 70-90% lower expression and activity of ICMT present improvement in body weight, grip strength

and bone structure, and exhibit extended survival when compared with Zmpste24-/-Icmt+/+ littermates.

This diminished activity of ICMT leads to mislocalization of prelamin A in the nucleus and activates

AKT-mammalian target of rapamycin (mTOR) signaling, which in turn delays cell senescence.

However, these findings are not completely in accord with the results of Cao et al. and Graziotto et al.,

who showed that treatment with the mTOR inhibitor rapamycin activates autophagic clearance of

progerin and reduces nuclear abnormalities (84, 85). These results clearly show that ICMT and mTOR

are implicated in premature aging, but the precise mechanism and relationship between them awaits

further investigation.

I.6. Mechanisms underlying CVD in progeria

I.6.1. VSMC loss

Progressive VSMC loss was reported both in HGPS patients (20-22) and in various progeria

mouse models (58, 59, 61), indicating its importance in the pathogenesis of the premature vascular

disease. Depletion of VSMCs in the media has also been described in physiological aging (86),

although not as severe as is seen in HGPS.

Page 31: ATHEROSCLEROSIS IN PROGERIA

I. INTRODUCTION

20

VSMCs in the blood vessels are subjected to high mechanical stress related to the blood flow.

In normal conditions, cells respond to increased shear stress by increasing the expression of A-type

lamins and changing their distribution within the nucleus (63, 87, 88). This response to physical stress

is perturbed in progerin-expressing cells (81, 89) and may lead to cell damage and death. This premise

is supported by the work of Verstraeten et al., showing reduced viability and increased apoptosis of

HGPS fibroblasts under repetitive mechanical strain (90). One possible explanation for the progerin-

induced increase in mechanosensitivity relates to changes in the expression of proteins controlling

cytoskeleton organization, mechanotransduction and extracellular matrix production (91, 92). Song et

al. showed that the ascending aorta of G608G BAC transgenic mice exhibits reduced expression of

vimentin (91), a cytoskeletal protein attached to the nucleus, endoplasmic reticulum, and mitochondria

that is important for maintaining cellular integrity (93). This correlation between downregulation of

mechanotransduction proteins and high shear stress in progerin-expressing VSMCs might partially

explain their loss in HGPS.

To elucidate how progerin leads to VSMC death, various groups established an in vitro model

of human smooth muscle cells (SMCs) differentiated from HGPS induced pluripotent stem cells

(iPSCs). Liu et al. showed that iPSC-derived progerin-expressing SMCs exhibit premature senescence

associated with vascular aging, and they identified DNA-dependent protein kinase catalytic subunit

(DNA-PKcs) as a binding partner of progerin (94). DNA-PKcs, which is a catalytic subunit of a nuclear

DNA-PK, participates in the non-homologous end joining (NHEJ) pathway of DNA repair. Recently,

Kinoshita et al. analyzed the interactome of lamin A mutant forms and they found that progerin, unlike

wild-type lamin A, cannot bind to proteins related to DNA damage response, including DNA-PK

holoenzyme (95). They also reported that progerin expression in VSMCs, but not in endothelial cells

(ECs), induces DNA-PK activation and growth arrest, leading to cell senescence. Given that the results

of Liu et al. and Kintoshita et al. are somewhat contradictory, the interaction between progerin and

DNA-PK and its subunits should be further defined.

Zhang et al. found that HGPS iPSC-derived SMCs exhibit severe proliferative defects triggered

by a caspase-independent mechanism (96). They also reported that progerin expression in SMCs is

associated with inhibition of poly(ADP-ribose) polymerase 1, an important regulator of DNA repair,

and induces the activation of the error-prone NHEJ response. The subsequent prolonged mitosis results

in mitotic catastrophe, leading to SMC death. Similar to progerin, prelamin A induces DNA damage

and increases DNA damage response in aged VSMCs (97, 98). This response might be the consequence

of impaired recruitment of 53 binding protein-1 (53BP1) to the sites of DNA damage due to a defective

Page 32: ATHEROSCLEROSIS IN PROGERIA

I. INTRODUCTION

21

import of 53BP1 to the nucleus, related to mislocalization of nucleoporin 153 (99). Defective DNA

damage repair has been also described in non-vascular HGPS cells and in progeria mouse models (100-

102). All of the above findings confirm the contribution of DNA damage response to progerin-driven

VSMC death. Interestingly, DNA damage plays an important role in normal aging (103).

I.6.2. Vascular calcification

HGPS patients exhibit excessive aortic and valvular calcification (20, 31, 32, 104), which is

also a hallmark of physiological aging (105). Likewise, vascular calcification is observed in progeroid

transgenic G608G BAC and knock-in LmnaG609G/+ mice (58, 60). Villa-Bellosta et al. (60) found

abnormally high expression of the osteogenic markers bone morphogenetic protein 2 (Bmp2) and Run-

related transcription factor-2 (Runx2) in calcified aortas from LmnaG609G/+ mice, without alterations in

the anti-calcification agents matrix Gla-protein and fetuin A. Moreover, LmnaG609G/+-derived primary

VSMCs showed a reduced capacity to inhibit calcium deposition in vitro, which was associated with a

decreased amount of extracellular inorganic pyrophosphate (ePPi), the major endogenous inhibitor of

vascular calcification. The diminished level of ePPi was related to an impairment in ePPi synthesis due

to decreased ATP production (the main substrate for ePPi synthesis) and an upregulation of tissue-

nonspecific alkaline phosphatase (the main enzyme involved in PPi hydrolysis) and ectonucleoside

triphosphatase diphosphohydrolase 1 (the enzyme that hydrolyzes ATP to release Pi). Accordingly,

plasma concentrations of ePPi and ATP were found to be lower in LmnaG609G/+ mice than in Lmna+/+

littermates. Moreover, injection of exogenous PPi prevented vascular calcification in LmnaG609G/G609G

mice.

The diminished capacity of progerin-expressing VSMCs to prevent calcification might be

related to a switch from a contractile to an osteochondrocytic phenotype. This notion is supported by

the work of Liu et al., who showed that prelamin A accumulation in VSMCs interferes with DNA

damage repair leading to osteogenic differentiation (98). These findings are of special interest because,

similar to progerin, prelamin A accumulates with age in medial VSMCs and atherosclerotic lesions,

and has been proposed as a novel biomarker of VSMC aging (97). Prelamin A activates DNA damage-

related ataxia telangiectasia mutated /ataxia telangiectasia and Rad3-related signaling, and induces

senescence-associated secretory phenotype in VSMCs (98). Prelamin A-expressing senescent VSMCs

release pro-calcification factors, such as BMP2, which may trigger calcification both locally and at

remote sites.

Page 33: ATHEROSCLEROSIS IN PROGERIA

I. INTRODUCTION

22

A recent study also demonstrated that VSMCs cultured in calcifying medium show higher

expression of lamin A and prelamin A, accompanied by upregulated gene expression of relevant

regulators of calcification, such as Runx2, osteocalcin and osteopontin, and increased calcium

deposition (106). Remarkably, human mesenchymal stems cells expressing progerin also show elevated

levels of osteopontin and exhibit enhanced osteogenic differentiation (107). In summary, lamin A and

its mutant or unprocessed forms participate in osteoblastic differentiation and vascular calcification,

indicating a further need to examine the role of progerin and prelamin A in premature and physiological

aging.

I.6.3. Endothelial dysfunction

EC dysfunction is an essential event in atherosclerosis development, which is the life-

threatening component of HGPS. It has been well established that aortic regions subjected to turbulent

blood flow and high shear stress, such as the ascending aorta, are more susceptible to atheroma plaque

formation (108, 109). ECs can sense and respond to different types of blood flow. Song et al. observed

that ECs in the ascending aorta of G608G BAC mice form an intact monolayer in the zones with an

almost complete loss of VSMCs (91). Moreover, these ECs showed a greater than 8-fold increase in

vimentin levels compared with ECs in zones with preserved VSMCs. An elevated level of vimentin in

progerin-expressing ECs might augment their resistance to increased shear stress, thus explaining the

presence of well-preserved endothelium in HGPS vessels (20).

A recent study showed that accumulation of prelamin A in ECs, through blocking lamin A

maturation, induces cell senescence and promotes intercellular adhesion molecule 1-dependent

monocyte adhesion to ECs (110). Since monocyte adhesion to ECs and extravasation is an important

step in the initiation of the atheroma plaque formation, further studies are needed to unravel the

relationship between progerin and prelamin A expression in ECs and atheroma build-up.

I.6.4. Cardiac electrical alterations

Previous studies in HGPS patients revealed repolarization abnormalities, such as ST segment

depression/elevation and negative and biphasic T waves, which were especially evident at advanced

stages of the disease (17, 30). Progeriod Zmpste24-/- mice show similar alterations manifesting as T-

wave flattening (30). Although progerin-expressing LmnaG609G/G609G and prelamin A-expressing

Page 34: ATHEROSCLEROSIS IN PROGERIA

I. INTRODUCTION

23

Zmpste24-/- mice develop severe bradycardia as they age, HGPS patients present cardiac rhythm that

remains within the normal range; yet, the heart rate seemed to be slower in older patients (30, 59).

Moreover, 18-20-week-old Zmpste24-/- animals showed prolonged PQ interval and QRS complex,

indicating defective cardiac conduction (30). These alterations were accompanied by mislocalization

of the gap junction protein connexin 43, which was also evident in heart tissue from HGPS patients

(30). Intercellular connectivity defects might therefore underlie cardiac electrical defects in progeria.

Taken together, these findings suggest that cardiac alterations in HGPS patients and progeroid mice are

a characteristic of progeria that could increase the risk of arrhythmias and lead to premature death.

I.7. Endoplasmic reticulum (ER) stress and the unfolded protein response (UPR)

Aging has been linked to the capacity of an organism to cope with stress stimuli (111, 112).

Organisms have a wide variety of stress response mechanisms that can act at the cellular or organelle-

specific level to restore homeostasis, including heat shock response, autophagy, mitochondrial and ER

stress responses, remodeled proteasome, and the DNA damage response (112). These stress responses

involve recognition of the damage, transmission of the stress signal to the nucleus, production of stress-

related proteins and their translocation to the site of damage. Impaired responses to stress are believed

to contribute to age-related tissue damage, but compromised stress response can also accelerate aging.

Alterations in the ER stress response have been linked to some age-associated diseases such as

diabetes, heart disease and neurodegenerative disorders (113-115). In response to certain stress stimuli,

such as free cholesterol, oxidized lipids, high glucose, mitochondrial dysfunction and calcium

imbalance, unfolded proteins accumulate within the ER, leading to ER stress (116, 117). In an attempt

to reestablish homeostasis, ER stress triggers several adaptive mechanisms, which together are known

as the UPR (116, 118). UPR activation leads to diminished misfolded or unfolded protein load by

reducing the influx of proteins into the ER, synthesizing proteins responsible for folding and quality

control in the ER, and increasing the ER membrane size (119-122). However, under persistent stress

conditions, UPR fails to restore homeostasis and triggers programmed cell death (123).

Three classes of ER stress sensors are known to mediate UPR activation: inositol-requiring

enzyme 1 (IRE1), protein kinase RNA-like ER kinase (PERK), and activating transcription factor 6

(ATF6) (118) (Fig. 2). These transmembrane receptors are maintained in an inactive form through

binding to the ER chaperone 78 kDa glucose-regulated protein (GRP78), also known as

Page 35: ATHEROSCLEROSIS IN PROGERIA

I. INTRODUCTION

24

immunoglobulin heavy chain-binding protein. In the presence of high levels of misfolded or unfolded

proteins, GRP78 dissociates from IRE-1, PERK and ATF6, enabling their activation (124).

During the early stages of the UPR, protein synthesis is inhibited by blocking translation in a

process mediated by the PERK-dependent phosphorylation of eukaryotic translation initiator factor 2α

(eIF2α) (125). Moreover, regulated IRE1-dependent decay leads to degradation of specific mRNAs

that code for ER-located proteins (126-128). ER stress also stimulates autophagy though the IRE1-JUN

N-terminal kinase pathway, in order to eliminate damaged ER and aberrant protein aggregates (129).

Generally, these first-line responses aim to reduce protein influx into the ER and facilitate adaptive and

repair mechanisms that restore homeostasis.

During the middle stages of the UPR, the transcription factors ATF6 fragment (ATF6f), spliced

X box-binding protein 1 (XBP1s) and ATF4 are activated to restore ER function and promote survival.

Under ER stress, ATF6 dissociates from GRP78 and translocates to the Golgi apparatus, where it is

cleaved (130, 131). The resulting cytosolic fragment ATF6f enters the nucleus and stimulates

transcription of the ER-associated protein degradation (ERAD) constituents and XBP1 (132, 133).

Dimerization and autotransphosphorylation of IRE1 upon ER stress results in the activation of

its cytosolic RNase domain (134). Active IRE1 excises 26 nucleotides from the mRNA coding for

XBP1, leading to a shift in the reading frame and the production of an active and stable transcription

factor called XBP1s (s for “spliced”) (133, 135, 136). Subsequently, XBP1s enters the nucleus and

activates the transcription of genes that encode proteins involved in ERAD and protein folding (137,

138). XBP1s also regulates phospholipid synthesis, leading to the ER membrane expansion upon ER

stress (139). By contrast, XBP1, which is the protein product of unspliced XBP1 mRNA, is unstable

and inhibits UPR target gene transcription (135).

Although PERK activation leads to the attenuation of translation, it upregulates the expression

of some proteins, including ATF4 and GRP78 (140, 141). ATF4 regulates genes responsible for amino

acid metabolism, redox balance, protein folding and autophagy (122, 142). Moreover, it also promotes

eIF2α dephosphorylation and translation recovery by increasing the expression of growth arrest and

DNA damage-inducible gene 34 (143).

Although UPR is a pro-survival mechanism, it can lead to cell death in situations of acute or

chronic ER stress. The central factor orchestrating the ER stress-related apoptosis is DNA damage-

inducible transcript 3 protein (DDIT3), also called growth arrest and DNA damage-inducible protein

or CCAAT/enhancer-binding protein homologous protein (144). Under normal conditions, the amount

Page 36: ATHEROSCLEROSIS IN PROGERIA

I. INTRODUCTION

25

of DDIT3 in the cell is low; however, the protein level of DDIT3 increases via IRE1-, PERK- and

ATF6- pathways upon ER stress activation (although ATF4 plays the main role in the activation of

DDIT3 gene expression). In turn, DDIT3 upregulates tumor necrosis factor-related apoptosis-inducing

ligand (TRAIL) receptors, leading to activation of the extrinsic apoptotic pathway (145). In addition to

DDIT3, other factors mediating ER stress-related apoptosis have been proposed, including IRE1 and

caspase 12 (122).

Figure 2. A simplified scheme of endoplasmic reticulum (ER) stress and the unfolded protein response (UPR)

activation. Three UPR-related molecules are activated upon ER stress: activating transcription factor 6 (ATF6), inositol-

requiring enzyme 1 (IRE1), and protein kinase RNA-like ER kinase (PERK). At the early stages of the UPR, protein

translation is diminished through PERK-mediated phosphorylation of eukaryotic translation initiator factor 2α (eIF2α).

Moreover, initiation of regulated IRE1- dependent decay (RIDD) leads to decay of some mRNAs. In parallel, autophagy

is activated via the IRE1α–JUN N-terminal kinase (JNK) pathway. At the middle stages of the UPR, three transcription

factors (activating transcription factor 6 cytosolic fragment (ATF6f), spliced X box-binding protein 1 (XBP1s) and

ATF4) promote various adaptive responses in order to reestablish ER function and promote cell survival. However,

unresolved ER stress triggers apoptosis via different pathways, with DNA damage-inducible transcript 3 protein

(DDIT3; also known as C/EBP-homologous protein) being the key transcription factor regulating this process. Dashed

lines indicate events promoting apoptosis. ERAD: ER-associated protein degradation.

Page 37: ATHEROSCLEROSIS IN PROGERIA

27

II. OBJECTIVES

Page 38: ATHEROSCLEROSIS IN PROGERIA

II. OBJECTIVES

29

Since the discovery in 2003 of the HGPS-causing mutation in the LMNA gene, much progress

has been made in unraveling the mechanisms underlying progerin-induced premature aging and death.

However, few studies have investigated the molecular alterations triggered by progerin accumulation

in VSMCs, major players in atherosclerosis development (60, 94-96, 146). Moreover, the mechanisms

through which progerin accelerates atherosclerosis remain largely undefined, in part due to the lack of

adequate mouse models.

Ongoing HGPS clinical trials are testing FTIs, either as monotherapy or in combination with

bisphosphonates and statins (75-77). These strategies are clearly not curative since most HGPS

symptoms persist in treated patients, including CVD and premature death. Further research is therefore

needed to identify new disease mechanisms and to develop more efficient therapies to inhibit

atherosclerosis development, the main cause of death in HGPS patients.

The main objectives of this thesis were the following:

1. To generate the first progeroid mouse model exhibiting progerin-induced acceleration of

atherosclerosis that phenocopies the main vascular alterations observed in HGPS patients.

2. To assess the relative contribution of macrophages and VSMCs to progerin-driven

atherosclerosis.

3. To identify the molecular mechanisms underlying progerin-induced atherosclerosis.

4. To test new treatments to prevent cardiovascular events and extend lifespan of progeric

mice that could be used in HGPS patients.

Page 39: ATHEROSCLEROSIS IN PROGERIA

31

III. MATERIALS AND METHODS

Page 40: ATHEROSCLEROSIS IN PROGERIA

III. MATERIALS AND METHODS

33

III.1. Mice

All experimental and other scientific procedures with animals conformed to EU Directive

2010/63EU and Recommendation 2007/526/EC, enforced in Spanish law under Real Decreto 53/2013.

Animal protocols were approved by the local ethics committees and the Animal Protection Area of the

Comunidad Autónoma de Madrid (PROEX76/14, PROEX78/14, PROEX167/16).

All mice used in this study were male on the C57BL/6J genetic background. Apolipoprotein E-

deficient (Apoe-/-, The Jackson Laboratory, stock no: 002052), LmnaG609G/+ (59), LmnaLCS/+ (59),

SM22αCre (Tagln-Cre, The Jackson Laboratory, stock no: 017491), and LysMCre (147) mice were

purchased or kindly provided by collaborators. These lines were used to generate atherosclerosis-

susceptible mouse models with ubiquitous progerin expression (Apoe-/-LmnaG609G/G609G), VSMC-

specific progerin expression (Apoe-/-LmnaLCS/LCSSM22αCre), macrophage (myeloid)-specific progerin

expression (Apoe-/-LmnaLCS/LCSLysMCre), and their corresponding controls expressing normal lamin

A/C (Apoe-/-Lmna+/+) or lamin C only (Apoe-/-LmnaLCS/LCS). Moreover, to compare the atherosclerosis-

prone (Apoe-/-) and atherosclerosis-resistant (Apoe+/+) backgrounds, we bred LmnaLCS/LCSSM22αCre,

LmnaLCS/LCS, LmnaG609G/G609G and Lmna+/+ mice. With the exception of longevity studies, animals on

the Apoe-/- background were sacrificed at 8, 16, 21-23, 27 or 51 weeks of age (indicated in the figure

legends). LmnaLCS/LCSSM22αCre and LmnaLCS/LCS mice were sacrificed at 38 week of age.

III.2. Longevity study

Beginning at 4 weeks of age, animals were weighed and inspected for health and survival at

least once per week (checks were more frequent for Apoe-/-LmnaG609G/G609G mice). Diseased animals

were examined by a specialized veterinarian blinded to genotype. Animals that met humane endpoint

criteria were euthanized and the deaths recorded. Animals sacrificed due to hydrocephalus,

malocclusion, inter-male aggression or other reasons unconnected to phenotype, were excluded from

the analysis (normally at a very early stage of the study).

III.3. High-fat diet (HFD) experiments

For diet-induced atherosclerosis studies, animals were maintained for 8 weeks on a HFD (10.7%

total fat, 0.75% cholesterol, S9167-E010, Ssniff), beginning at 8 weeks of age. Mice were sacrificed at

16 weeks of age after an overnight fast.

Page 41: ATHEROSCLEROSIS IN PROGERIA

III. MATERIALS AND METHODS

34

III.4. Hematology and serum biochemical analysis

Animals were fasted overnight for all blood analyses, which were performed by specialized

staff from the CNIC Animal Unit. For hematology, blood samples were collected in Microvette 100

EDTA tubes (Sarstedt) and analyzed using a PENTRA 80 hematology analyzer (Horibo). For

biochemical analysis, blood samples were collected in plastic tubes, incubated at room temperature

(RT) for 2-3 hours to allow clotting, and centrifuged at 2000 × g for 5 minutes. Serum was stored at -

80°C until samples from all experiments were collected. Because of volume limitations, serum samples

were pooled from approximately 2-3 animals of the same experimental group. Specimens with overt

hemolysis were excluded from testing. Biochemical variables were analyzed using a Dimension RxL

Max Integrated Chemistry System (Siemens Healthineers).

III.5. Blood pressure measurement

Blood pressure and heart rate measurements were performed in conscious mice using the

BP2000 noninvasive automated tail-cuff system (Visitech Systems). All experiments were performed

in the morning to avoid variability related to circadian oscillations in the blood pressure (148, 149).

Animals were trained during five consecutive days (first week) and then experimental data was

collected during 5 consecutive days (second week). For each mouse, at least 10 measurements of blood

pressure and heart rate were registered each day. Final measurements were preceded by 10 preliminary

measurements to allow the animals to settle. Values that were equal to 0 (which arose from equipment

error or animal movements) were excluded from the analysis. For each day, the median was calculated

for the heart rate, systolic and diastolic blood pressure. The mean from 5 days was used for further

analysis.

III.6. ECG

Mice were anesthetized with 1.5-2% isoflurane and four ECG electrodes were inserted

subcutaneously into the limbs. ECG was recorded in the morning (to account for circadian variations)

using the MP36R system (Biopac Systems). ECG data were analyzed using AcqKnowledge software

by specialized staff blinded to the genotype.

Page 42: ATHEROSCLEROSIS IN PROGERIA

III. MATERIALS AND METHODS

35

III.7. Treatment

Tauroursodeoxycholic acid (TUDCA, 580549, Calbiochem) was dissolved in phosphate-

buffered saline (PBS) and passed through a 0.22 µm filter. After genotyping, mice were randomized

into treatment and control groups (an equal or similar number of animals from each litter was assigned

to each group). TUDCA (400 mg/kg) or PBS was administered intraperitoneally 3 times a week,

beginning at 6 weeks of age in the case of Apoe-/-LmnaG609G/G609G mice and at 8 weeks of age in the

case of Apoe-/-LmnaLCS/LCSSM22αCre mice. Atherosclerosis experiments included PBS- or TUDCA-

treated Apoe-/-Lmna+/+ and Apoe-/-LmnaLCS/LCS control mice.

III.8. Quantification of atherosclerosis burden

Mouse aortas were fixed with 4% formaldehyde/PBS, cleaned of fatty tissue, and stained with

0.2% Oil Red O (ORO, O0625, Sigma). The thoracic aorta or/and aortic arch was then incised

longitudinally and pinned out flat, intimal side up, for computer-assisted planimetric analysis. Images

were taken with a digital camera (OLYMPUS UC30) mounted on a stereo microscope (OLYMPUS

SZX3). The percentage of lesion area (ORO-stained) was quantified using SigmaScan Pro 5 software

(Systat Software Inc.) by an observer blinded to genotype.

III.9. Histology and immunofluorescence

For all mice on the Apoe-/- background, formaldehyde-fixed aortic arches were cleaned of fatty

tissue, incubated in 30% sucrose in PBS overnight at 4°C, and embedded in Tissue-Tek® OCT

compound (SAKURA, Netherlands) for cryostat sectioning. Serial 8-μm sections were stained with

ORO, hematoxylin-eosin (H&E), and Masson’s trichrome. For immunofluorescence, sections were

blocked and permeabilized for 1 hour at RT in PBS containing 0.3% Triton X-100 (9002-93-1, Sigma),

5% bovine serum albumin (BSA, A7906, Sigma), and 5% normal goat serum (005-000-001, Jackson

ImmunoResearch). Next, sections were incubated overnight at 4°C with the following antibodies: anti-

smooth muscle α-actin (Sma-Cy3, C6198, Sigma, 1:200), anti-CD68 (MCA1957, Serotec, 1:200), and

anti-progerin/lamin A (sc-20680, Santa Cruz, 1:100) diluted in PBS containing 0.3% Triton X-100 and

2.5% normal goat serum. Samples were incubated with corresponding secondary antibodies (Alexa

Fluor, Invitrogen) and the nucleic acid stain Hoechst 33342 (B2261, Sigma) for 2 hours at RT, and

mounted using Fluoromount G imaging medium (4958-02, Affymetrix eBioscience).

Page 43: ATHEROSCLEROSIS IN PROGERIA

III. MATERIALS AND METHODS

36

Aortic arches of LmnaLCS/LCSSM22αCre and LmnaLCS/LCS mice were fixed in 4% formaldehyde

in PBS, cleaned of fatty tissue, dehydrated to xylene and embedded in paraffin. Thoracic aortas used

for atherosclerosis burden quantification (from TUDCA-treatment experiments) were also embedded

in paraffin. Serial 4-μm sections were deparaffinized, rehydrated, and stained with H&E, and Masson’s

trichrome. For immunofluorescence, antigen retrieval was performed using 10 mM sodium citrate

buffer (pH 6). Then, samples were blocked with PBS containing 5% BSA and 5% normal goat serum

for 1 hour at RT. Sections were incubated for 2 hours at RT with an anti-smooth muscle α-actin

antibody (Sma-Cy3, C6198, Sigma, 1:200) and Hoechst 33342 stain diluted in PBS with 2.5% normal

goat serum, and mounted using Fluoromount G imaging medium.

Mouse hearts were fixed in 4% formaldehyde in PBS, divided into two parts (apex and upper

part containing aortic root with aortic valve), dehydrated to xylene and embedded in paraffin. Heart

(apex) sections from 6 different levels were prepared. The aortic root was sectioned throughout the

aortic valve. Serial 4-μm sections were deparaffinized, rehydrated, and stained with H&E, and

Masson’s trichrome.

ORO-, H&E- and Masson’s trichrome-stained sections were scanned with a NanoZoomer-RS

scanner (Hamamatsu), and images were exported using NDP.view2. Immunofluorescence images were

acquired with the Zeiss LSM 700 confocal microscope. Images were analyzed using NDP.view2 and

ImageJ Fiji software by an observer blinded to genotype. Aortic media and adventitia thickness,

collagen and lipid medial content, and VSMC content were analyzed in approximately 3 sections (for

aortic arch) and/or 4 (for thoracic aorta) per animal, and the mean was used for the statistical analysis.

Atherosclerotic plaque area, plaque collagen and VSMC content were quantified in 3 different zones

of the aortic valve (beginning, middle, and end) per animal, and the mean was used for the statistical

analysis.

III.10. Sample collection and preparation for RNAseq

Eight-week-old mice (Apoe-/-LmnaG609G/G609G, Apoe-/-Lmna+/+, Apoe-/-LmnaLCS/LCSSM22αCre

and Apoe-/-LmnaLCS/LCS) were sacrificed by CO2 inhalation, and thoracic aortas were extracted, cleaned

of fatty tissue and digested with 2 mg/ml collagenase (CLS-2, Worthington) for 10 minutes at 37°C to

separate medial and adventitial tissue. Medial aortas from 3-4 mice of the same genotype were pooled

and snap frozen. Samples were disrupted using TissueLyser (Qiagen), and total RNA was isolated with

Page 44: ATHEROSCLEROSIS IN PROGERIA

III. MATERIALS AND METHODS

37

QIAzol (Qiagen). RNA integrity was confirmed by RNA electrophoresis and with an Agilent 2100

Bioanalyzer.

III.11. RNAseq library preparation, sequencing, and generation of FastQ files

Total RNA (500 ng) was used to generate barcoded RNAseq libraries using the TruSeq RNA

Sample Preparation Kit v2 (Illumina). Briefly, poly A+ RNA was purified using poly-T oligo-attached

magnetic beads through two rounds of purification, followed by fragmentation and first and second

cDNA strand synthesis. Next, cDNA 3’ ends were adenylated and the adapters were ligated, followed

by PCR library amplification. Finally, library size was checked using the Agilent 2100 Bioanalyzer

DNA 1000 chip and concentration was determined in a Qubit® fluorometer (Life Technologies).

Libraries were sequenced on a HiSeq2500 sequencer (Illumina) to generate 60-base single reads. FastQ

files for each sample were obtained using CASAVA v1.8 (Illumina). NGS experiments were performed

in the CNIC Genomics Unit. RNAseq data were deposited in the NCBI SRA, accession number:

SRP099105.

III.12. Differential expression analysis

Sequencing reads were pre-processed by means of a pipeline that used FastQC (150) to assess

read quality, and Cutadapt (151) to trim sequencing reads and eliminate Illumina adaptor sequences,

and to discard reads that were shorter than 30 base pairs. The number of reads obtained per sample was

in the range of 8 to 14 million. The resulting reads were mapped against the mouse transcriptome

(GRCm38, release 76; aug2014 archive) and quantified using RSEM v1.17 (152). The percentage of

aligned reads was in the range of 79 to 82%. Data were then processed with a differential expression

analysis pipeline that used the Bioconductor package EdgeR (153) for normalization and differential

expression testing. Only genes with at least 1 count per million in at least 4 samples (13,664 genes)

were considered for statistical analysis. Three comparisons were made to identify differentially

expressed genes in our models: 1) Apoe-/-LmnaG609G/G609G vs Apoe-/-Lmna+/+; 2) Apoe-/-

LmnaLCS/LCSSM22αCre vs Apoe-/-LmnaLCS/LCS; and 3) Apoe-/-LmnaLCS/LCS vs Apoe-/-Lmna+/+. The lists of

genes detected as differentially expressed in the 3 comparisons were subjected to a series of

bioinformatic analyses. The list of 240 genes shared between comparisons 1 and 2 was extracted and

the logFC values (base-2 logarithm of fold change) for those genes were plotted against each other

Page 45: ATHEROSCLEROSIS IN PROGERIA

III. MATERIALS AND METHODS

38

using Microsoft Excel to check for correlation. Differential expression analysis was performed in the

CNIC Bioinformatics Unit. Area-proportional Venn diagrams were generated using BioVenn (154) to

visualize the overlap between data sets.

III.13. Pathway analysis

Ingenuity Pathway Analysis (IPA, Qiagen) was used for more comprehensive RNAseq data

analysis. Briefly, core analyses were performed for the 3 comparisons to visualize pathways altered by

progerin expression as well as by the lack of lamin A. Benjamini-Hochberg correction of the P value

was applied to extract the most significant pathways and stacked bar charts were exported. A

comparison analysis was performed to compare results obtained with ubiquitous and VSMC-specific

progeroid models. Heatmaps showing canonical pathways and upstream regulators were exported. For

more detailed information about IPA tools (Global Canonical Pathways, Upstream regulators and

Comparative analysis), see www.ingenuity.com.

III.14. RNA extraction and cDNA preparation

Tissues were homogenized using TissueLyser (Qiagen), and total RNA was extracted with

QIAzol reagent (Qiagen). The RNA pellet was dissolved in RNase-free water and concentration was

measured in a NanoDrop spectrophotometer (Wilmington). RNA (2 µg) was transcribed to cDNA using

the High Capacity cDNA Reverse Transcription Kit (Applied Biosystems).

III.15. PCR detection of lamin A and progerin

Lamin A and progerin mRNA levels were quantified according to a protocol adapted from Yang

et al. (155). cDNA (100 ng for medial aortas and 200 ng for other organs) was amplified by PCR using

DNA polymerase (Biotools, Spain). PCR products were separated on a 2% agarose gel containing

ethidium bromide. Images were taken with a Molecular Imager® Gel Doc™ XR+ System (BioRad)

and analyzed with Image Lab™ (BioRad).

Page 46: ATHEROSCLEROSIS IN PROGERIA

III. MATERIALS AND METHODS

39

III.16. Quantitative real-time PCR

qPCR reactions were prepared using Power SYBR® Green PCR Master Mix (Applied

Biosystems). PCR reaction mixes were loaded on 384-well plates (Applied Biosystems) and run on a

7900-FAST-384 thermal cycler (Applied Biosystems). All reactions were performed in triplicate.

Primers used in this study are as follows: Calr (F: 5′-CCAGAAATTGACAACCCTGAA-3′, R: 5′-

CCTTAAGCCTCTGCTCCTCAT-3′), Ddit3 (F: 5′-ATATCTCATCCCCAGGAAACG-3′, R: 5′-

CTCCTGCTCCTTCTCCTTCAT-3′), Dnajb9 (F: 5′-AGAATTAATCCTGGCCTCCAA-3′, R: 5′-

GGCATCCGAGAGTGTTTCATA-3′), Hspa5 (F: 5′-GTGGGAGGAGTCATGACAAAA-3′, R: 5′-

TTCAGCTGTCACTCGGAGAAT-3′), Hsp90b1 (F: 5′-AGTGGAAGAGGACCTGGGTAA-3′, R: 5′-

AGCGAGTGCATTTTCATCAGT-3′), Pdia4 (F: 5′-TCCTGAAGGATGGAGATGATG-3′, R: 5′-

ACCTGGGCTCATACTTGGACT-3′), Gusb (F: 5′-GAGTATGGAGCAGACGCAATC-3′, R: 5′-

TCCGACCACGTATTCTTTACG-3′), and Hprt (F: 5′-AGGCCAGACTTTGTTGGATTT-3′, R: 5′-

GGCTTTGTATTTGGCTTTTCC-3′).

III.17. Statistical analysis

Experimental data are presented as mean (error bars indicate standard error of the mean (SEM))

for parametric data, or median with interquartile range (error bars indicate minimum and maximum)

for non-parametric data. For small sample sizes, data were plotted as independent points (dot plots).

Based on all experiments, distribution of a variable (lesion size, nucleus count, etc.) was assessed using

the Kolmogorov-Smirnov and D'Agostino-Pearson normality tests. If the distribution was normal in

most of the experiments, a two-tailed t-test was used, except for the validation of RNAseq data by

qPCR and VSMC content quantification after TUDCA treatment where a one-tailed t-test was used. If

the distribution of a variable was skewed, the two-tailed Mann-Whitney test was used, except for the

adventitia-to-media ratio quantification after TUDCA treatment where a one-tailed Mann-Whitney test

was used. To compare multiple groups, one-way ANOVA with Tukey´s post hoc test was used for both

parametric and non-parametric data because this approach is resistant to normality violations with

similar sized groups. A log-rank (Mantel-Cox) test was used for Kaplan-Meier survival curves.

Differences were considered significant at P < 0.05. Statistical analysis was performed with GraphPad

Prism 5.

Page 47: ATHEROSCLEROSIS IN PROGERIA

41

IV. RESULTS AND DISCUSSION

Page 48: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

43

IV.1. Ubiquitous progerin expression aggravates atherosclerosis in HFD-fed Apoe-/- mice

Among the available HGPS-like mouse models, LmnaG609G/G609G knock-in mice ubiquitously

expressing progerin recapitulate most of the clinical manifestations of HGPS, including growth

impairment, bone abnormalities, lipodystrophy, vascular calcification, and reduced survival (average

lifespan: ≈15 weeks) (59, 60). However, we did not observe atherosclerosis in aortas of LmnaG609G/G609G

mice even when 8-week-old animals were challenged with HFD for 8 weeks (Fig. 3). This finding is

consistent with the observation that, unlike humans, mice are extremely resistant to atherosclerosis

development, due in part to differences in cholesterol and lipoprotein metabolism (156).

Figure 3. LmnaG609G/G609G mice do not show any signs of atherosclerosis development. LmnaG609G/G609G and Lmna+/+

mice were fed high-fat diet for 8 weeks starting at 8 weeks of age. Mice were sacrificed at 16 weeks (close to their maximum

survival) and aortas were stained with Oil Red O to visualize lipid-rich atheroma plaques. No red staining was observed

indicating absence of atherosclerosis.

To circumvent this limitation, we generated an atherosclerosis-prone mouse model of HGPS by

crossing LmnaG609G/+ mice with Apoe-/- mice, a widely-used model of atherosclerosis (156). As

expected, Apoe-/-LmnaG609G/G609G mice showed decreased body weight and a shortened life span

(median survival: 18.5 weeks) as compared with Apoe-/-Lmna+/+ littermates with normal Lmna gene

expression (median survival: 117.6 weeks) (Fig. 4). Notably, the Apoe-/-LmnaG609G/G609G phenotype,

including survival and body weight curves, was similar to that of LmnaG609G/G609G mice (59).

Page 49: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

44

Figure 4. Ubiquitous progerin expression in Apoe-/-

LmnaG609G/G609G

mice impairs postnatal growth and reduces

lifespan. (A) Postnatal body weight curves for Apoe-/-

Lmna+/+

mice (n=7) and Apoe-/-

LmnaG609G/G609G

mice (n=14). (B)

Kaplan-Meier survival curves. Median survival = 18.5 weeks for Apoe-/-

LmnaG609G/G609G

mice (n=14) and 117.6 weeks for

Apoe-/-

Lmna+/+

mice (n=7); P<0.001. (C) Representative photograph of 16-week-old males. Data are presented as mean ±

SEM in A. Statistical analysis was performed by log-rank test in B.

To study the influence of progerin on atherosclerosis development, 8-week-old mice were

challenged for 8 weeks with HFD. Pre-HFD fasting serum lipid levels, including total cholesterol, free

cholesterol, LDL and HDL, were indistinguishable between experimental groups (Fig. 5A). However,

post-HFD levels were significantly lower in Apoe-/-LmnaG609G/G609G mice than in Apoe-/-Lmna+/+

controls (Fig. 5B), probably due to a reduced food intake related to premature aging and associated

disease progression in progeric mice. Nevertheless, despite the lower serum lipid levels, atherosclerosis

burden assessed by ORO staining was 1.8-fold higher in the aortic arch and 3.1-fold higher in the

thoracic aorta of HFD-fed Apoe-/-LmnaG609G/G609G mice (Fig. 5C). Both groups had focal aortic lesions,

but the aortic surface of progeroid Apoe-/-LmnaG609G/G609G mice was covered with lipid deposits, which

were not seen in control Apoe-/-Lmna+/+ mice (Fig. 5C).

Page 50: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

45

Figure 5. Ubiquitous progerin expression in Apoe-/-

LmnaG609G/G609G

mice accelerates atherosclerosis upon high-fat

diet (HFD). Mice were fed HFD for 8 weeks starting at 8 weeks of age. (A) Fasting serum pre-HFD levels of total

cholesterol, free cholesterol, low-density lipoprotein (LDL), and high-density lipoprotein (HDL) in 8-week-old Apoe-/-

LmnaG609G/G609G mice (n=5) and Apoe-/-Lmna+/+ mice (n=7). (B) Fasting serum post-HFD levels of total cholesterol, free

cholesterol, LDL, and HDL in 16-week-old Apoe-/-LmnaG609G/G609G mice (n=6) and Apoe-/-Lmna+/+ mice (n=8). (C)

Representative aortic arches (top) and thoracic aortas (middle and bottom) stained with Oil Red O; graphs show

quantification of atherosclerosis burden in Apoe-/-

LmnaG609G/G609G

mice (n=6 aortic arches; n=12 thoracic aortas) and Apoe-

/-Lmna

+/+ mice (n=9 aortic arches; n=13 thoracic aortas). Data are shown as mean ± SEM in A and B, and as median with

interquartile range and minima and maxima in C. Statistical differences were analyzed by two-tailed t-test in A and B and

by two-tailed Mann-Whitney test in C. *P<0.05, **P<0.01, ***P<0.001.

Histology revealed that this abnormality was due to excessive lipid accumulation in the aortic

tunica media in atheroma-free zones of Apoe-/-LmnaG609G/G609G aorta (Fig. 6A, B). These lipid deposits

typically corresponded to the medial regions, with an almost complete loss of VSMCs (Fig. 6A). As in

human HGPS arteries, VSMC depletion in Apoe-/-LmnaG609G/G609G mice was accompanied by increased

collagen content in the media and abnormal elastin structure (Fig. 6A, B). Importantly, Apoe-/-

LmnaG609G/G609G mice developed adventitial thickening and inflammation (Fig. 6A, B), a relevant

feature of HGPS. Some of the pathologies described above have been observed in other progeroid

Page 51: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

46

mouse models, such as VSMC depletion in knock-in LmnaG609G/G609G and transgenic G608G BAC mice

(58, 59), but not as early and as severe as in the model described here (Fig. 6C).

Figure 6. Ubiquitous progerin expression in Apoe-/-

LmnaG609G/G609G

mice produces severe vascular pathology,

including lipid retention in the media, vascular smooth muscle cell (VSMC) loss, adventitial thickening, and

increased collagen content upon high-fat diet (HFD). Mice were fed HFD for 8 weeks starting at 8 weeks of age. (A)

Representative staining of aorta sections with Oil Red O (ORO), hematoxylin & eosin (H&E), and Masson’s Trichrome.

(B) Graphs show quantification of lipid content in atheroma-free zones of the media (as % of ORO-positive area), adventitia-

to-media thickness ratio, and collagen content in the media (% of blue staining); n=6-8. Scale bar, 200 µm. (C)

Representative immunofluorescence images of aortas stained with anti-smooth muscle actin (Sma) antibody (red) and

Hoechst3442 (blue); graphs show quantification of VSMC content in the media as either % of Sma-positive area (top) or

nucleus count (bottom); n=6-8. Scale bar, 50 µm. Box and whisker plots in B show medians, interquartile range, and minima

and maxima; data in C are mean ± SEM. Statistical analysis was performed by two-tailed Mann-Whitney test in B, and

two-tailed t-test in C. **P<0.01, ***P<0.001. m: media, a: adventitia.

Apoe-/-LmnaG609G/G609G mice also showed significant hematological alterations in leukocyte

subpopulations, including lower pre- and post-HFD lymphocyte and monocyte counts (Fig. 7 and 8).

Since hematological analysis is based on the size and granularity of the cell, some of these

measurements may be compromised because progerin affects nuclear shape. These findings should

Page 52: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

47

therefore be confirmed by flow cytometry using specific cell surface marker antibodies to precisely

define changes in circulating leukocyte subpopulations of Apoe-/-LmnaG609G/G609G mice.

Although HGPS patients have increased platelet count (17), we did not find significantly higher

amount of platelets in the blood of Apoe-/-LmnaG609G/G609G mice as compared with Apoe-/-Lmna+/+

controls. This progerin-driven alteration could be masked by the Apoe deficiency, because

atherosclerosis-resistant LmnaG609G/G609G mice with an intact Apoe gene have a higher platelet count

than Lmna+/+ littermates (data not shown).

Figure 7. Apoe-/-LmnaG609G/G609G mice have lower leukocyte, lymphocyte and monocyte counts pre-high fat diet (HFD).

Pre-HFD (fasting) hematology results for 8-week-old Apoe-/-LmnaG609G/G609G mice (n=18) and Apoe-/-Lmna+/+ mice (n=18).

Data are shown as median with interquartile range and minima and maxima. Statistical differences were analyzed by two-

tailed Mann-Whitney test. ***P<0.001.

Page 53: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

48

Figure 8. Apoe-/-LmnaG609G/G609G mice show higher neutrophil and lower leukocyte, lymphocyte, monocyte, and

eosinophil counts post-high fat diet (HFD). Post-HFD (fasting) hematology results for 16-week-old Apoe-/-

LmnaG609G/G609G mice (n=17) and Apoe-/-Lmna+/+ mice (n=17). Data are shown as median with interquartile range and

minima and maxima. Statistical differences were analyzed by two-tailed Mann-Whitney test. **P<0.01, ***P<0.001.

In this study, we have generated the first mouse model of progerin-induced acceleration of

atherosclerosis, a pathological process that leads to myocardial infarction- or stroke-related death in

most children with HGPS. As our Apoe-/-LmnaG609G/G609G mice faithfully recapitulate most of the

aspects of the human disease, they are a valuable tool to investigate the molecular and cellular

mechanisms underlying premature CVD and death in HGPS.

IV.2. VSMC-specific progerin expression in HFD-fed Apoe-/- mice aggravates atherosclerosis

and shortens lifespan

Atherosclerosis is a complex process involving many cell types, including VSMCs and

macrophages (157). Immunohistopathological characterization of atherosclerotic plaques in HGPS

patients suggests that both cell types contribute to progerin-driven atherogenesis (20). Since Apoe-/-

LmnaG609G/G609G mice showed medial VSMC depletion and decreased numbers of circulating

monocytes (which give rise to macrophages in atheromata), we generated mice with specific progerin

expression in VSMCs or macrophages to study the relative contribution of these cell types in progeria.

Page 54: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

49

To this end, we used knock-in LmnaLCS/LCS (LaminC-STOP) mice expressing only lamin C (59).

The LmnaLCS allele consists of a modified Lmna gene in which a neomycin resistance gene flanked

with two loxP sequences was introduced after exon 10 to abolish lamin A production (Fig. 9).

Additionally, an HGPS-equivalent point mutation (c.1827C>T; p.G609G) was inserted in exon 11.

Excision of the neomycin resistance cassette in the presence of Cre recombinase enables progerin

production (as well as lamin C and some residual lamin A). LmnaLCS/LCS mice expressing lamin C and

lacking lamin A are apparently normal, but are slightly heavier and longer-lived than wild-type

controls, and show decreased energy metabolism (158).

Figure 9. Schematic representation of LmnaLCS allele before and after Cre-mediated recombination. The LmnaLCS

allele carries an HGPS-causing mutation (p.G609G, indicated with an asterisk) in exon 11. However, the only product of

the LmnaLCS allele is lamin C due to the insertion of a neomycin resistance gene after exon 10 which disables production of

lamin A. In the presence of Cre recombinase, the neomycin resistance cassette, which is flanked with loxP sequences, is

excised and three proteins are produced via alternative splicing: lamin C, progerin and lamin A.

We first performed pilot studies to assess whether the absence of lamin A may aggravate

atherosclerosis. To do this, we generated Apoe-/-LmnaLCS/LCS mice and analyzed atheroma plaque

formation in 16-week-old mice challenged for 8 weeks with HFD (beginning at 8 weeks of age). No

Page 55: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

50

significant differences in atherosclerosis burden, serum lipid levels, and hematological parameters were

found between control Apoe-/-Lmna+/+ and Apoe-/-LmnaLCS/LCS mice (Fig. 10).

Figure 10. Fat-fed Apoe-/-LmnaLCS/LCS mice expressing only lamin C do not show more atherosclerosis than Apoe-/-

Lmna+/+ control mice expressing wild-type lamin A/C. Mice were fed high-fat diet (HFD) for 8 weeks starting at 8 weeks

of age. (A) Representative examples of aortas stained with Oil Red O and quantification of atherosclerosis burden in Apoe-

/-LmnaLCS/LCS mice (n=8 aortic arches, n=11 thoracic aortas) and Apoe-/-Lmna+/+ mice (n=7 aortic arches, n=11 thoracic

aortas). (B) Post-HFD fasting serum levels of total cholesterol, free cholesterol, low-density lipoprotein (LDL), and high-

density lipoprotein (HDL) in Apoe-/-LmnaLCS/LCS (n=6) and Apoe-/-Lmna+/+ (n=7) mice. (C) Post-HFD (fasting) hematology

results for 16-week-old Apoe-/-LmnaLCS/LCS mice (n=11) and Apoe-/-Lmna+/+ mice (n=11). Data are shown as median with

interquartile range and minima and maxima in A and C and as mean ± SEM in B. Statistical differences were analyzed by

two-tailed Mann-Whitney test in A and C and by two-tailed t-test in B.

Page 56: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

51

The above result allowed Apoe-/-LmnaLCS/LCS mice to serve as controls for the cell-type-specific

models Apoe-/-LmnaLCS/LCSLysMCre (macrophage-specific) and Apoe-/-LmnaLCS/LCSSM22Cre (VSMC-

specific). Progerin expression was verified by immunofluorescence in atheroma-containing aorta and

by PCR in other organs. These studies confirmed that control Apoe-/-LmnaLCS/LCS mice did not express

progerin in any organ tested (Fig. 11 and 12). By contrast, Apoe-/-LmnaLCS/LCSSM22Cre mice

abundantly expressed progerin in medial VSMCs and to a lesser extent in the adventitia (Fig. 12), as

well as in heart (Fig. 11). Progerin expression in Apoe-/-LmnaLCS/LCSLysMCre mice was detected in

intimal macrophages (Fig. 12), but was either absent or negligible in other organs (Fig. 11).

Figure 11. Progerin expression in different organs of progeroid and control mice. Representative images of PCR

products showing lamin A and progerin mRNA expression in liver, kidney, spleen, and heart of 8-week-old mice of the

indicated genotypes. Lamin A but not progerin was expressed in all organs from Apoe-/-

Lmna+/+

mice, and progerin was

detected in all organs from Apoe-/-

LmnaG609G/G609G

mice (which also expressed low levels of lamin A). Lamin A and progerin

were undetectable or negligible in all organs of Apoe-/-

LmnaLCS/LCS

, Apoe-/-

LmnaLCS/LCS

SM22αCre, and Apoe-/-

LmnaLCS/LCS

LysMCre mice, with the exception of Apoe-/-

LmnaLCS/LCS

SM22αCre heart, which expressed both progerin and

lamin A. Arbp was used as an endogenous control.

Page 57: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

52

Figure 12. Progerin is expressed specifically in vascular smooth muscle cells (VSMCs) of Apoe-/-

LmnaLCS/LCSSM22αCre aorta and in macrophages of Apoe-/-LmnaLCS/LCSLysMCre aorta. Representative images of

atheroma-containing aortas from 16-week-old mice fed a high-fat diet for 2 months starting at 8 weeks of age. Progerin was

visualized in white, macrophages were detected with anti-CD68 antibody (green), and VSMCs with anti-Sma antibody

(red). Nuclei were stained with Hoechst3442 (blue). Scale bar, 50µm. Amplified images show VSMC-rich media and a

macrophage-rich atherosclerotic plaque.

Page 58: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

53

Apoe-/-LmnaLCS/LCSLysMCre mice with macrophage-specific progerin expression showed no

significant differences in body weight, longevity and organ size relative to controls (Fig. 13 and 14).

Apoe-/-LmnaLCS/LCSSM22Cre mice with VSMC-specific progerin expression appeared normal early in

life, but stopped gaining weight after approximately 20 weeks of age and died at a median age of 34.3

weeks (Fig. 13 and 14).

Figure 13. Vascular smooth muscle cell (VSMC)-specific progerin expression in Apoe-/-

LmnaLCS/LCS

SM22αCre mice

reduces lifespan. (A) Postnatal body weight curves for Apoe-/-

LmnaLCS/LCS

mice (n=9), Apoe-/-

LmnaLCS/LCS

SM22αCre mice

(n=12), and Apoe-/-

LmnaLCS/LCS

LysMCre mice (n=6). The weight curve for Apoe-/-

LmnaLCS/LCS

SM22αCre mice is shown up

to the time when approximately 90% of the animals were dead. Data are presented as mean ± SEM. (B) Kaplan-Meier

survival curves. Median survival = 34.3 weeks for Apoe-/-

LmnaLCS/LCS

SM22αCre mice (n=17), 106.3 weeks for Apoe-/-

LmnaLCS/LCS

mice (n=9), and 116.3 weeks for Apoe-/-

LmnaLCS/LCS

LysMCre mice (n=6). P<0.0001 for Apoe-/-

LmnaLCS/LCS

SM22αCre mice vs Apoe-/-

LmnaLCS/LCS

controls. Statistical analysis was performed by log-rank test. (C)

Representative photograph of 16-week-old males.

Page 59: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

54

Figure 14. Apoe-/-

LmnaLCS/LCS

SM22αCre and Apoe-/-

LmnaLCS/LCS

LysMCre mice, unlike Apoe-/-LmnaG609G/G609G, show

normal organ size. Representative images of liver, kidney, spleen, and heart of 16-week-old mice of the indicated

genotypes. Scale bar 1 cm.

We next studied atherosclerosis development in the cell type-specific models following the

same protocol as that used for the ubiquitous progerin-expressing Apoe-/-LmnaG609G/G609G mice. The

three experimental groups showed no inter-group differences in fasting serum lipid levels or

hematological parameters, either pre- or post-HFD (Fig. 15 and 16).

Figure 15. Apoe-/-LmnaLCS/LCSSM22αCre and Apoe-/-LmnaLCS/LCSLysMCre mice have normal serum cholesterol levels.

Mice were fed high-fat diet (HFD) for 8 weeks starting at 8 weeks of age. (A) Pre-HFD fasting serum levels of total

cholesterol, free cholesterol, low-density lipoprotein (LDL), and high-density lipoprotein (HDL) in 8-week-old Apoe-/-

LmnaLCS/LCS mice (n=5), Apoe-/-LmnaLCS/LCSSM22αCre mice (n=4), and Apoe-/-LmnaLCS/LCSLysMCre mice (n=6). (B) Post-

HFD fasting serum levels of total cholesterol, free cholesterol, LDL, and HDL in 16-week-old Apoe-/-LmnaLCS/LCS mice

(n=10), Apoe-/-LmnaLCS/LCSSM22αCre mice (n=7), and Apoe-/-LmnaLCS/LCSLysMCre mice (n=7). Data are shown as mean ±

SEM. Statistical differences were analyzed by one-way ANOVA with Tukey´s post hoc test.

Page 60: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

55

Figure 16. Apoe-/-LmnaLCS/LCSSM22αCre and Apoe-/-LmnaLCS/LCSLysMCre mice show normal hematological

parameters. Mice were fed high-fat diet (HFD) for 8 weeks starting at 8 weeks of age. (A) Pre-HFD (fasting) hematology

results for 8-week-old Apoe-/-LmnaLCS/LCS mice (n=17), Apoe-/-LmnaLCS/LCSSM22αCre mice (n=11), and Apoe-/-

LmnaLCS/LCSLysMCre mice (n=11). (B) Post-HFD (fasting) hematology results for 16-week-old Apoe-/-LmnaLCS/LCS mice

(n=17), Apoe-/-LmnaLCS/LCSSM22αCre mice (n=11), and Apoe-/-LmnaLCS/LCSLysMCre mice (n=11). Data are shown as median

with interquartile range and minima and maxima. Statistical differences were analyzed by one-way ANOVA with Tukey´s

post hoc test.

Page 61: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

56

Atherosclerosis burden in the thoracic aorta was similar between Apoe-/-LmnaLCS/LCSLysMCre

and Apoe-/-LmnaLCS/LCS mice. By contrast, lesion burden in Apoe-/-LmnaLCS/LCSSM22αCre mice was 3.7-

fold higher (Fig. 17A). Moreover, histological analysis of Apoe-/-LmnaLCS/LCSSM22αCre mice revealed

the same marked aortic phenotype as observed in the ubiquitous progeroid model, including massive

VSMC loss, adventitial thickening, elastin structure changes, and medial lipid accumulation (Fig. 17B,

C).

Figure 17. Vascular smooth muscle cell (VSMC)-specific progerin expression in fat-fed Apoe-/-

LmnaLCS/LCS

SM22αCre

mice triggers severe vascular pathology, including atherosclerosis, VSMC loss, lipid retention in the media and

adventitial thickening. Mice were fed high-fat diet (HFD) for 8 weeks starting at 8 weeks of age. (A) Representative

examples of thoracic aortas stained with Oil Red O (ORO); the graph shows quantification of atherosclerosis burden in

Apoe-/-

LmnaLCS/LCS

mice (n=24) Apoe-/-

LmnaLCS/LCS

SM22αCre mice (n=17), and Apoe-/-

LmnaLCS/LCS

LysMCre mice (n=19).

(B) Representative staining of aorta sections with ORO and hematoxylin & eosin (H&E); graphs show quantification of

lipid content in atheroma-free zones of the media (% of ORO-positive area) and adventitia-to-media thickness ratio; n=6-

8. Scale bar, 50 µm. (C) Representative immunofluorescence images of aortas stained with anti-smooth muscle actin (Sma)

antibody (red) and Hoechst3442 (blue); graphs show quantification of VSMC content in the media as either % of Sma-

positive area (top) or nucleus count (bottom); n=6-8. Scale bar, 50 µm. Box and whisker plots in A and B show medians,

interquartile range, and minima and maxima; data in C are mean ± SEM. Statistical analysis was performed by one-way

ANOVA with Tukey´s post hoc test in A, B, and C. ***P<0.001, m: media, a: adventitia.

Page 62: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

57

The above results provide the first direct evidence that restricting progerin expression to

VSMCs is sufficient to accelerate atherosclerosis and reduce lifespan, offering the possibility to study

CVD independently of other premature aging symptoms. Another advantage of Apoe-/-

LmnaLCS/LCSSM22αCre animals is the lack of overt growth defects, which may permit diagnostic

techniques and interventional strategies that are unattainable in the undersized Apoe-/-LmnaG609G/G609G

model.

IV.3. Progerin triggers plaque vulnerability

Previous studies have shown that loss of VSMCs in the tunica media may lead to changes in

the composition and stability of atherosclerotic plaques (159). We therefore performed a more in-depth

analysis of atheromata in the aortic root of Apoe-/-LmnaG609G/G609G and Apoe-/-LmnaLCS/LCSSM22αCre

mice and their respective controls. Three different zones of the aortic root were analyzed, at the

beginning, in the middle, and at the end of the aortic valve. Consistent with our previous results in the

aorta (Fig. 5 and 17), we observed a higher atherosclerosis burden in both progeria models than in the

respective controls, and this was accompanied by fibrosis and inflammation of the adjacent cardiac

tissue (Fig. 18A, B). Moreover, a greater percentage of the aortic surface was affected by the

atherosclerosis process in both Apoe-/-LmnaG609G/G609G and Apoe-/-LmnaLCS/LCSSM22αCre mice (Fig.

18A, B). Importantly, all Apoe-/-LmnaG609G/G609G and Apoe-/-LmnaLCS/LCSSM22αCre mice exhibited

coronary VSMC loss (Fig. 18C, D), and some of them developed coronary atherosclerosis (data not

shown). In addition, we observed aortic valve degeneration and plaque formation on the aortic valve

leaflets in Apoe-/-LmnaG609G/G609G and Apoe-/-LmnaLCS/LCSSM22αCre mice, pathologies that were absent

in control mice (Fig. 18A, B, and data not shown).

Page 63: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

58

Figure 18. Fat-fed Apoe-/-

LmnaG609G/G609G

and Apoe-/-

LmnaLCS/LCS

SM22αCre mice exhibit increased atherosclerosis

in the aortic root together with inflammation of the adjacent cardiac tissue and coronary alterations (including

vascular smooth muscle cell (VSMC) loss and thickening and inflammation of the adventitia). Mice were fed high-

fat diet (HFD) for 8 weeks starting at 8 weeks of age. Serial sections of the aortic root were made and 3 different regions

were stained with Masson’s Trichrome. (A, B) Representative photographs of atheroma plaque in the aortic root of

Apoe-/-

LmnaG609G/G609G

(A) and Apoe-/-

LmnaLCS/LCS

SM22αCre mice (B), and their corresponding controls; graphs show

quantification of plaque area, with each point representing mean from three different aortic root regions (n=5). Scale

bar, 200 µm. Dashed lines indicate medial perimeter (the last layer of elastin), dotted lines indicate the luminal surface

of the atheroma plaque. (C, D) Representative photographs of coronary artery of Apoe-/-

LmnaG609G/G609G (C) and Apoe

-/-

LmnaLCS/LCS

SM22αCre mice (D), and their corresponding controls. Scale bar, 100 µm. Data in A and B are shown as

mean ± SEM. Statistical analysis was performed by two-tailed t-test in A and B. **P<0.01, ***P<0.001.

Importantly, Apoe-/-LmnaG609G/G609G and Apoe-/-LmnaLCS/LCSSM22αCre atheromata showed

features of vulnerable plaques. Accordingly, plaques of Apoe-/-LmnaG609G/G609G and Apoe-/-

LmnaLCS/LCSSM22αCre mice contained erythrocytes, indicating healed plaque fissure (Fig. 19A, B,

top), and some animals showed hemorrhage in the aortic valve itself (Fig. 19A, bottom). We also found

evidence of thrombus formation (Fig. 19B, bottom), which might be related to plaque erosion.

Page 64: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

59

Figure 19. Atherosclerotic plaques of fat-fed Apoe-/-

LmnaG609G/G609G

and Apoe-/-

LmnaLCS/LCS

SM22αCre mice show

evidence of disruption. Mice were fed high-fat diet (HFD) for 8 weeks starting at 8 weeks of age. Serial sections of the

aortic root were made and 3 different regions were stained with Masson’s Trichrome. (A) An example of intraplaque (top)

and intravalve hemorrhage (bottom) in Apoe-/-

LmnaG609G/G6090G

mice. (B) An example of intraplaque hemorrhage (top) and

thrombus formation (bottom) in Apoe-/-

LmnaLCS/LCS

SM22αCre mice.

IV.4. Progerin expression accelerates atherosclerosis in Apoe-/- mice fed normal chow

To gain insight into the kinetics of progerin-induced atherogenesis and to assess the role of

cholesterol in this process, we performed experiments in mice fed normal chow. Histological

examination of aortas from young, 8-week-old mice revealed no differences between the progeroid

models and their controls, with tissue almost free of atherosclerosis and no obvious structural

alterations (Fig. 20A, C, E). By contrast, atherosclerosis was evident in the thoracic aorta of 16-week-

old Apoe-/-LmnaG609G/G609G and Apoe-/-LmnaLCS/LCSSM22Cre mice, as well as regions with VSMC loss

and increased collagen content (Fig. 20B, D, E). Notably, Apoe-/-LmnaLCS/LCSSM22Cre mice

presented a marginally more pronounced vascular phenotype than Apoe-/-LmnaG609G/G609G mice, with

lipid retention in the media and adventitial thickening (data not shown), probably due to normal food

intake and the consequent cholesterol level similar to that of the control mice.

Page 65: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

60

Figure 20. Vascular disease in 16-week-old but not 8-week-old Apoe

-/-Lmna

G609G/G609G and Apoe

-/-

LmnaLCS/LCS

SM22αCre mice fed normal chow. (A-D) Representative images of thoracic aortas stained with Oil Red O

and quantification of atherosclerosis burden in mice of the indicated ages and genotypes. Controls for Apoe-/-

LmnaG609G/G609G

and Apoe-/-

LmnaLCS/LCS

SM22αCre mice were Apoe-/-

Lmna+/+

and Apoe-/-

LmnaLCS/LCS

, respectively. The number of mice is

indicated below each graph. (E) Representative histology sections of Masson´s Trichrome-stained aortic arches extracted

from mice of the indicated ages and genotypes fed normal chow diet. White arrowheads indicate regions with vascular

smooth muscle cell loss. Scale bar, 50 µm. Data in A-D are shown as median with interquartile range and minima and

maxima. Statistical differences were analyzed by the two-tailed Mann-Whitney test. *P<0.05, **P<0.01.

Page 66: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

61

Our findings in both the ubiquitous and the VSMC-specific progeric mouse models fed either

normal chow or HFD indicate that VSMCs are particularly sensitive to progerin accumulation, which

leads to cell damage and death that is accelerated in the presence of other stress stimuli, including high

cholesterol (Fig. 21). Mechanical stress also seems to play an important role in this process because

VSMC loss differs between distinct aortic regions and positively correlates with high shear stress (e.g.,

VSMC loss is higher in the aortic arch than in the thoracic aorta, data not shown). Dead VSMCs are

replaced by extracellular matrix components, which retain lipids and lipoproteins. Additionally, pro-

inflammatory signals from dead cells may activate the vascular endothelium and enhance its

permeability for the circulating lipoprotein particles. Cell debris and lipid/lipoprotein deposits trigger

inflammation and thickening of the adventitia and accelerate atheroma plaque formation. Finally,

vulnerable plaque disruption may lead to cardiovascular events, such as myocardial infarction, and

cause premature death.

Figure 21. Proposed mechanism of vascular smooth muscle cell (VSMC)-specific progerin toxicity leading to

accelerated atherosclerosis and premature death. Progerin expression triggers VSMC death, which is accelerated in the

presence of other stress stimuli, such as high cholesterol or high mechanical stress. Dead VSMCs are replaced with

extracellular matrix, which retains more lipids and lipoproteins. This may create a vicious circle – the more lipids are

accumulated in the media, the more VSMCs die (and vice versa). Cell death and lipid/lipoprotein deposits drive

inflammation and thickening of the adventitia and accelerate atherosclerosis. Finally, vulnerable plaques lead to death,

probably due to a cardiovascular event, such as myocardial infarction.

Page 67: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

62

IV.5. Progerin expression in VSMCs leads to progressive hypotension

The ubiquitous and VSMC-specific models of progeria presented a very similar aortic

phenotype, both leading to aortic luminal dilatation and plaque buildup. We therefore hypothesized

that progerin expression might affect blood pressure. To address this, mice were fed normal chow and

blood pressure was measured at approximately 16 weeks of age during 5 consecutive days. Blood

pressure was not different between Apoe-/-LmnaG609G/G609G mice and Apoe-/-Lmna+/+ controls (Fig.

22A); however, Apoe-/-LmnaG609G/G609G mice developed severe bradycardia (Fig. 22A), which has been

previously reported for progeroid LmnaG609G/G609G and Zmpste24-/- mice (30, 59). Furthermore, diastolic

blood pressure in Apoe-/-LmnaLCS/LCSSM22Cre mice, which showed more pronounced vasculopathy,

was significantly lower than in Apoe-/-LmnaLCS/LCS littermates (Fig. 22B). Follow-up of the Apoe-/-

LmnaLCS/LCSSM22Cre animals showed a further significant decline in both systolic and diastolic blood

pressure at 26 weeks of age (Fig. 22C).

These results conflict with findings in progeria patients. Specifically, two different studies

reported an increased incidence of hypertension in HGPS children (17, 23); however, those results

could be considered biased. In the first work by Merideth et al. only the highest blood pressure readings

were reported, although this parameter was measured 6 times during the course of the study (17). It is

puzzling why the authors elected to report and analyze only the maximum blood pressure values instead

of the mean or median, raising concerns about the validity of their conclusions. The second study by

Gerhard-Herman et al. reported that blood pressure values were increased in around 30% of the patients

as compared with age-matched controls (23). When height-age standards were applied to account for

the growth impairment, systolic and diastolic blood pressures were higher in approximately half of the

patients. Nevertheless, the authors did not report how many normotensive and hypotensive children

were in the studied cohort. They also did not provide extensive details about experimental design, for

example, the number of times that blood pressure was measured and how the data were analyzed. In

addition, some published reports describing HGPS phenotypes include as supplementary information

all the blood pressure measurements registered during the study and many of these values are below

the normal range for age-matched controls (20). Based on these observations in patients and our

findings in mice, more studies are warranted to elucidate the effect of progerin on blood pressure in

children with HGPS, with special emphasis on hypotension and occasional hypertension incidence.

Page 68: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

63

Figure 22. Apoe-/-LmnaLCS/LCSSM22Cre mice develop progressive hypotension while Apoe-/-LmnaG609G/G609G mice

display blood pressure and heart rate variability. Blood pressure and heart rate measurements were performed in

conscious 16-week-old (for both progeria models) and 26-week-old (for VSMC-specific progeria model) mice fed normal

chow diet. (A, B, C) Heart rate, systolic and diastolic blood pressure analysis for Apoe-/-

LmnaG609G/G609G

, Apoe-/-

LmnaLCS/LCS

SM22αCre, Apoe-/-

Lmna+/+

and Apoe-/-

LmnaLCS/LCS

mice. (C, D, E) Variability in the heart rate, systolic and

diastolic blood pressure for Apoe-/-

LmnaG609G/G609G

, Apoe-/-

LmnaLCS/LCS

SM22αCre, Apoe-/-

Lmna+/+

and Apoe-/-

LmnaLCS/LCS

mice measured as coefficient of variation (CV). n=10-13. Data are presented as mean ± SEM. Statistical analysis

was performed by two-tailed t-test. *P<0.05, **P<0.01, ***P<0.001.

Although Apoe-/-LmnaG609G/G609G mice, unlike Apoe-/-LmnaLCS/LCSSM22Cre mice, did not

develop prominent hypotension until 16 weeks of age, we observed a substantial instability and

variability in heart rate and blood pressure (Fig. 22D-F). Blood pressure variability is defined as

changes in blood pressure over time (hours, days, weeks, months etc.), whereas instability refers to

transient oscillations in blood pressure, typically in response to pain, stress or change in posture, and

adds to total variability. Variability and instability in blood pressure in humans is associated with higher

risk of vascular events (such as stoke) related to plaque rupture (26, 27). Thus, blood pressure instability

and variability in Apoe-/-LmnaG609G/G609G mice could be linked to intraplaque hemorrhages observed in

Page 69: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

64

those animals upon HFD feeding (see subsection IV.3). However, intraplaque hemorrhages were also

present in HFD-fed Apoe-/-LmnaLCS/LCSSM22Cre mice, which do not show blood pressure variability

(at least at the same age). Since blood pressure measurements were performed in animals fed normal

chow presenting only moderate vasculopathy, the relationship between plaque disruption and blood

pressure variation in progeria mice should be further defined by preforming blood pressure analysis in

HFD-fed progeroid mice.

IV.6. Apoe-/-LmnaG609G/G609G mice develop arrhythmias

Blood pressure measurements in conscious Apoe-/-LmnaG609G/G609G mice revealed heart rate

instability and variability, suggesting cardiac involvement (Fig. 22D). Moreover, a previous study has

described electrical cardiac alterations in the Zmpste24-deficient (Zmpste24-/-) progeria mouse model

(30). Given this information, we performed ECG in both ubiquitous and VSMC-specific progeria

models. In accordance with the results from conscious animals (subsection IV.5), 16-week-old

anesthetized Apoe-/-LmnaG609G/G609G mice presented severe bradycardia (Fig. 23A) together with

prolonged QRS, QT and QTc intervals (Fig. 23A). Importantly, 3 out of 7 mice showed arrhythmias

(Fig. 23B), which might explain the blood pressure instability and variability described in the previous

subsection. This result is also consistent with the observation that HGPS patients show repolarization

abnormalities resulting in an increased risk of arrhythmias and premature death (30).

Page 70: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

65

Figure 23. Apoe-/-LmnaG609G/G609G mice develop cardiac electrical defects resulting in arrhythmias. Electrocardiogram

(ECG) was performed in isoflurane-anesthetized 16-week-old mice fed normal chow. (A) ECG parameters for Apoe-/-

LmnaG609G/G609G (n=5) and Apoe-/-Lmna+/+ (n=7) mice. (B) Representative images of ECG recording for Apoe-/-Lmna+/+ (left)

and Apoe-/-LmnaG609G/G609G (right) mice. Apoe-/-LmnaG609G/G609G mice exhibit bradycardia and arrhythmia. QRSH, PH, TH

indicate amplitude of QRS complex, PH and TH waves, respectively. Data are presented as mean ± SEM. Statistical analysis

was performed by two-tailed t-test. *P<0.05, **P<0.01, ***P<0.001.

In contrast to the findings with Apoe-/-LmnaG609G/G609G mice, 16-week-old Apoe-/-

LmnaLCS/LCSSM22Cre mice did not present any evident ECG alterations (Fig. 24A). However, follow-

up of these animals revealed low voltage of QRS complex at 26 weeks of age (Fig. 24B), which might

indicate loss of viable myocardial tissue, for example, due to myocardial infarction. Low QRS voltage

on the ECG is a marker of the severity of heart failure, and is a risk factor for adverse outcomes in

patients with this condition (160). Likewise, many of the Apoe-/-LmnaLCS/LCSSM22Cre mice exhibited

T wave fattening or inversion (Fig. 24B and data not shown), further supporting the notion that these

animals may have cardiac involvement as they age, probably associated with progressive

atherosclerosis. Indeed, acute myocardial ischemia in mice has been related to the inverted T wave on

Page 71: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

66

the ECG (161). Remarkably, studies in HGPS patients revealed ST depression/elevation and negative

and biphasic T waves, especially evident at advanced stages of the disease (17, 30).

Figure 24. Apoe-/-LmnaLCS/LCSSM22Cre mice show normal cardiac electrical function at 16 weeks of age and lower

voltage of QRS complex and T wave at 26 weeks of age compared with Apoe-/-LmnaLCS/LCS littermates.

Electrocardiogram (ECG) was performed in isoflurane-anesthetized 16- and 26-week-old mice fed normal chow diet. (A)

ECG parameters for 16-week-old Apoe-/-LmnaLCS/LCSSM22Cre (n=7) and Apoe-/-LmnaLCS/LCS (n=7) mice. (B) ECG

parameters for 26-week-old Apoe-/-LmnaLCS/LCSSM22Cre (n=8) and Apoe-/-LmnaLCS/LCS (n=7) mice. QRSH, PH, TH

indicate amplitude of QRS complex, PH and TH waves, respectively. Data are presented as mean ± SEM. Statistical analysis

was performed by two-tailed t-test. *P<0.05.

IV.7. Apoe-/-LmnaLCS/LCSSM22Cre die from atherosclerosis-related causes

Despite an almost identical vascular phenotype, ubiquitous and VSMC-specific atherosclerosis-

prone progeria models showed different median survival (18.5 weeks versus 34.3 weeks, respectively).

Apoe-/-LmnaLCS/LCSSM22Cre mice, which did not display any overt aging phenotype, died suddenly

between 26 and 66 weeks of age. Interestingly, LmnaLCS/LCSSM22αCre mice with an intact Apoe gene

also presented VSMC loss and aortic adventitial thickening (Fig. 25A), but did not develop

Page 72: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

67

atherosclerosis and had a normal life span (Fig. 25B). These findings point to progerin-driven

atherosclerosis as the main cause of death in Apoe-/-LmnaLCS/LCSSM22Cre mice. By contrast, Apoe-/-

LmnaG609G/G609G mice progressively lost fat tissue, developed bone problems and muscle weakness, and

died between 15 and 24 weeks of age, a median survival very similar to that of atherosclerosis-free

LmnaG609G/G609G mice. These results strongly suggest that Apoe-/-LmnaG609G/G609G mice and

LmnaG609G/G609G mice die from the same cause, which is probably independent of atherosclerosis. These

observations, together with distinct cardiac alterations present in both atherosclerosis-susceptible

progeria models, lead us to hypothesize that the cause of death is different in ubiquitous and VSMC-

specific progeria models, at least in the majority of the cases.

Figure 25. LmnaLCS/LCSSM22αCre mice without atherosclerosis-susceptible background show aortic vascular smooth

muscle cell loss and adventitial thickening but have normal lifespan. LmnaLCS/LCS mice were used as controls. (A)

Representative histology sections of aortic arches stained with hematoxylin & eosin (H&E) and Masson´s Trichrome. Mice

fed normal chow were sacrificed at 38 weeks of age. m: media, a: adventitia. Scale bar, 100 µm. (B) Kaplan-Meier survival

curves of LmnaLCS/LCSSM22αCre mice (n=8) and LmnaLCS/LCS mice (n=10). Statistical differences in B were analyzed using

the log-rank test.

To analyze cardiovascular alterations, which may reveal the cause of death, we collected aorta,

heart and brain from normal chow-fed Apoe-/-LmnaG609G/G609G and Apoe-/-LmnaLCS/LCSSM22Cre mice

at an age close to their maximum survival (21-23 weeks for ubiquitous and 51 weeks for VSMC-

specific model). Atherosclerosis burden in the aortic arch and thoracic aorta was significantly higher

in Apoe-/-LmnaG609G/G609G mice than in Apoe-/-Lmna+/+ controls (Fig. 26A). When Apoe-/-LmnaLCS/LCS

and Apoe-/-LmnaLCS/LCSSM22Cre mice were compared, Apoe-/-LmnaLCS/LCSSM22Cre mice showed

increased lesion formation in the thoracic aorta (Fig. 26B); however no significant differences were

found in the aortic arch (Fig. 26B), likely because this athero-prone aortic region is saturated with

Page 73: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

68

plaques at 51 weeks (1 year) of age. Importantly, vascular pathology was much more severe in Apoe-/-

LmnaLCS/LCSSM22Cre mice than in Apoe-/-LmnaG609G/G609G mice (Fig. 26C).

Figure 26. Apoe-/-LmnaLCS/LCSSM22Cre mice have a more severe vascular phenotype than Apoe-/-LmnaG609G/G609G,

both at ages close to their maximum survival. Mice were fed normal chow and sacrificed at 21-23 weeks of age (Apoe-/-

LmnaG609G/G609G and control Apoe-/-Lmna+/+ mice) and 51 weeks of age (Apoe-/-LmnaLCS/LCSSM22Cre and control Apoe-/-

LmnaLCS/LCS mice). (A) Representative aortas of Apoe-/-

LmnaG609G/G609G

and Apoe-/-

Lmna+/+

mice stained with Oil Red O

(ORO); graphs show quantification of atherosclerosis burden in the aortic arch and thoracic aorta; n=6. (B) Representative

aortas of Apoe-/-LmnaLCS/LCSSM22Cre and Apoe-/-LmnaLCS/LCS mice stained with ORO; graphs show quantification of

atherosclerosis burden in the aortic arch and thoracic aorta; n=5-6. (C) Higher magnification of ORO-stained thoracic aortas

of the indicated genotypes. Data are shown as median with interquartile range and minima and maxima. Statistical

differences were analyzed by two-tailed Mann-Whitney test. *P<0.05, **P<0.01.

We next performed a more detailed analysis of the atherosclerotic plaques in the aortic root.

Both models exhibited aortic valve degeneration characterized by loss of cells and fibrosis (data not

shown). Apoe-/-LmnaG609G/G609G plaques were generally immature with intraplaque hemorrhages, which

were absent in the age-matched controls (Fig. 27). Two out of six Apoe-/-LmnaG609G/G609G animals

showed signs of coronary atherosclerosis (data not shown). In Apoe-/-LmnaLCS/LCSSM22Cre mice,

Page 74: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

69

atheromas were mature, severely calcified, and many of them presented chondroid metaplasia (Fig.

27). The atheromas that were not calcified showed thin fibrous caps and large necrotic cores (Fig 27),

characteristics of vulnerable plaques. The majority of analyzed Apoe-/-LmnaLCS/LCSSM22Cre mice

developed coronary atherosclerosis (data not shown).

Figure 27. Apoe-/-LmnaLCS/LCSSM22Cre mice have more severe atherosclerosis in the aortic root than Apoe-/-

LmnaG609G/G609G, both at ages close to their maximum survival. Mice were fed normal chow and sacrificed at 21-23 weeks

of age (Apoe-/-LmnaG609G/G609G and control Apoe-/-Lmna+/+ mice) and 51 weeks of age (Apoe-/-LmnaLCS/LCSSM22Cre and

control Apoe-/-LmnaLCS/LCS mice). Representative aortic root sections of Apoe-/-

LmnaG609G/G609G

, Apoe-/-

Lmna+/+

, Apoe-/-

LmnaLCS/LCSSM22Cre and Apoe-/-LmnaLCS/LCS mice stained with Masson´s Trichrome. Middle and bottom panels show

higher magnification of atherosclerotic plaques. m: media, p: plaque.

We also performed histopathological evaluation of the heart. Apoe-/-LmnaG609G/G609G hearts were

characterized by cardiomyocyte vacuolization that was absent in Apoe-/-Lmna+/+, Apoe-/-

LmnaLCS/LCSSM22Cre and Apoe-/-LmnaLCS/LCS mice (Fig. 28). This feature was previously described

in progeroid Zmpste24-/- mice (43). Importantly, cardiomyocyte vacuolization is frequently observed

in the border zones of infarcted heart tissue and it is a marker of ischemic injury (162-164).

Page 75: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

70

Figure 28. Apoe-/-LmnaG609G/G609G hearts exhibit cardiomyocyte vacuolization. Mice fed normal chow were sacrificed at

21-23 weeks of age (close to their maximum survival). Photographs show hematoxylin & eosin-stained cardiac tissue of

Apoe-/- Lmna+/+ (left) and Apoe-/- LmnaG609G/G609G (right) mice. Scale bar 200 µm. Black arrows indicate examples of

cardiomyocyte vacuolization.

Furthermore, two out of six Apoe-/-LmnaG609G/G609G hearts showed coronary atherosclerosis

(Table 1) and a collagen scar in the septum, indicative of a non-fatal infarct (Fig. 29). Nevertheless,

no alterations were detected in half of the animals analyzed (Table 1), except for vacuolization of

cardiomyocytes.

Genotype Intimal

hyperplasia

Coronary

atherosclerosis

Perivascular

fibrosis

Interstitial

fibrosis Calcification

Apoe-/-Lmna+/+ 1/6 0/6 0/6 0/6 0/6

Apoe-/-LmnaG609G/G609G 3/6 2/6 0/6 2/6 0/6

Apoe-/-LmnaLCS/LCS 2/6 1/6 1/6 0/6 0/6

Apoe-/-LmnaLCS/LCSSM22Cre 4/5 4/5 5/5 5/5 2/5

Table 1. Apoe-/-LmnaLCS/LCSSM22Cre mice exhibit a more severe cardiac phenotype than Apoe-/-LmnaG609G/G609G both

at ages close to their maximum survival. Mice fed normal chow were sacrificed at 21-23 weeks of age for Apoe-/-

LmnaG609G/G609G (and control Apoe-/-Lmna+/+) and 51 weeks of age for Apoe-/-LmnaLCS/LCSSM22Cre (and control Apoe-/-

LmnaLCS/LCS) mice. Serial sections of the heart (at 6 different levels of the apex) were prepared and stained with hematoxylin

& eosin and Masson´s Trichrome. Cardiac pathology was assessed in 5-6 mice per genotype.

Page 76: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

71

Figure 29. Evidence of coronary atherosclerosis and myocardial infarction in Apoe-/- LmnaG609G/G609G mice. Mice fed

normal chow were sacrificed at 21-23 weeks of age (close to their maximum survival). Photographs show consecutive

sections of an Apoe-/- LmnaG609G/G609G heart stained with hematoxylin & eosin (H&E) and Masson´s trichrome, showing

coronary atherosclerosis and infarct in the septum. Scale bar 1mm. RV: right ventricle; S: septum; LV: left ventricle.

Examination of Apoe-/-LmnaLCS/LCSSM22Cre hearts revealed that the majority presented

perivascular and interstitial fibrosis, coronary atherosclerosis and intimal hyperplasia (Table 1). We

also found calcification of the coronary plaque in some of the animals analyzed (Table 1). These

pathological alterations in the heart suggest that Apoe-/-LmnaLCS/LCSSM22Cre animals suffer many

small infarcts during their lifetime, similar to HGPS patients. However, no evidence of stroke was

found in the brain of Apoe-/-LmnaG609G/G609G and Apoe-/-LmnaLCS/LCSSM22Cre models (data not

shown).

In summary, comparison of mice at ages close to their maximum survival revealed a more

severe cardiac phenotype and atherosclerotic disease in Apoe-/-LmnaLCS/LCSSM22Cre mice than in

Apoe-/-LmnaG609G/G609G mice. The histopathological findings in the aorta and heart support our

hypothesis that Apoe-/-LmnaLCS/LCSSM22Cre animals die of atherosclerosis complications, whereas

Apoe-/-LmnaG609G/G609G die of atherosclerosis-independent processes. Plaque disruption leading to an

infarct and subsequent death may occur in Apoe-/-LmnaG609G/G609G mice only if their survival is close to

Page 77: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

72

26 weeks, the age at which Apoe-/-LmnaLCS/LCSSM22Cre begin to die. While we cannot rule out the

possibility that Apoe-/-LmnaG609G/G609G mice die from heart failure, it is probably related to other

complications such as arrhythmias (see subsection IV.6).

IV.8. Progerin expression in VSMCs activates endoplasmic ER stress and the UPR

In an attempt to identify mechanisms underlying the acceleration of atherosclerosis induced by

progerin, we conducted a transcriptomic analysis of aortas from the ubiquitous and VSMC-specific

progeria models. To identify drivers of disease rather than secondary changes, we collected arteries

before the onset of overt disease. The appropriate age for collection was determined based on our

previous histopathological analysis in Apoe-/-LmnaG609G/G609G and Apoe-/-LmnaLCS/LCSSM22Cre mice

fed normal chow (subsection IV.4).

Based on these findings and because VSMCs appear to be a major target of progerin, we

performed RNA sequencing (RNAseq) on VSMC-rich media of disease-free aortas from 8-week-old

Apoe-/-LmnaG609G/G609G and Apoe-/-LmnaLCS/LCSSM22Cre mice and their corresponding controls (Apoe-

/-Lmna+/+ and Apoe-/-LmnaLCS/LCS, respectively) (Fig. 30A). Four pooled samples per genotype were

collected, and PCR analysis confirmed proper progerin and lamin A expression (Fig. 30B). Differential

expression analysis revealed 776 significantly altered genes in the ubiquitous progeroid model and 931

altered genes in the VSMC-specific model (Fig. 30C). Of these differentially-regulated genes, 240

were common to both comparisons and had a high correlation (R2≈8, Fig. 30D).

Page 78: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

73

Figure 30. Progerin expression in vascular smooth muscle cells (VSMCs) causes altered gene expression. (A) Sample

preparation for RNA sequencing (RNAseq). (B) PCR confirmation of proper expression of lamin A and progerin in pooled

medial aortas used for RNAseq. Arbp was used as endogenous control. (C) Bioinformatic analysis detected 776

differentially expressed genes in medial aortas from Apoe-/- LmnaG609G/G609G mice with ubiquitous progerin expression

compared with Apoe-/-Lmna+/+ control mice expressing wild-type lamin A/C and 931 genes in medial aortas from Apoe-/-

LmnaLCS/LCSSM22αCre mice with VSMC-specific progerin expression compared with Apoe-/-LmnaLCS/LCS control mice

expressing lamin C only. There were 176 genes differentially expressed between the two control groups. The Venn diagram

shows the overlap between sets of deferentially expressed genes identified in each of the 3 comparisons. (D) Correlation

between logarithms of fold change calculated for the 240 genes shared between the comparisons “ubiquitous progerin vs

wild-type lamin A/C” and “VSMC-specific progerin vs lamin C only“.

To specifically separate the effect of progerin production from the absence of lamin A, we

performed analysis of the two controls, which revealed 176 genes differentially expressed between

Apoe-/-LmnaLCS/LCS aortas (expressing lamin C only) and Apoe-/-Lmna+/+ aortas (expressing wild-type

lamin A/C) (Fig. 30C). However, there was hardly any overlap between the gene sets affected by

progerin production in Apoe-/-LmnaG609G/G609G and Apoe-/-LmnaLCS/LCSSM22Cre mice and those

influenced by the absence of lamin A in Apoe-/-LmnaLCS/LCS mice (Fig. 30C). Likewise, progerin and

the absence of lamin A affected different pathways (Fig. 31).

Page 79: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

74

Figure 31. Pathways affected by lack of lamin A do not overlap with those induced by progerin expression. Stacked

bar charts representing pathways significantly changed after applying the Benjamini-Hochberg correction for multiple

testing in 3 comparisons: (A) Apoe-/-LmnaG609G/G609G (ubiquitous progerin) vs Apoe-/-Lmna+/+ (ubiquitous lamin A and lamin

C), (B) Apoe-/-LmnaLCS/LCSSM22αCre (vascular smooth muscle cell (VSMC)-specific progerin) vs Apoe-/-LmnaLCS/LCS

(ubiquitous lamin C, no lamin A), and (C) Apoe-/-LmnaLCS/LCS (ubiquitous lamin C, no lamin A) vs Apoe-/- Lmna+/+

(ubiquitous lamin A and lamin C). The numbers of genes in each category (from the Ingenuity Pathway Analysis data base)

are indicated above the bars.

Page 80: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

75

Comparison analysis identified four pathways that were significantly altered in both the

ubiquitous and the VSMC-specific progeroid models: fibrosis, nuclear factor erythroid 2-like 2

(NRF2)-mediated oxidative stress, the ER stress response, and the UPR (Fig. 32, left: Canonical

pathways). We also examined the predicted activation status of upstream regulators based on the

expression of their target genes. This analysis revealed that the most differentially regulated factors

belong to the ER stress response and ER stress-related UPR, for example, XBP1, ATF4 and DDIT3

(Fig. 32, right: Upstream regulators).

Figure 32. Progerin expression in vascular smooth muscle cell (VSMC)-rich aortic media activates endoplasmic

reticulum (ER) stress and unfolded protein response (UPR). RNAseq results were analyzed using Ingenuity Pathway

Analysis: (left) canonical pathway heatmap, showing processes affected by progerin expression in VSMC-rich medial

aortas. Asterisk (*) indicates pathways which are significantly changed in both comparisons after applying the Benjamini-

Hochberg correction for multiple testing; (right) upstream regulator heatmap, showing predicted activation states of

transcriptional regulators (black boxes indicate key molecules involved in ER stress and UPR regulation).

Page 81: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

76

To validate the RNAseq results, we performed quantitative real-time PCR on selected ER stress

response and UPR genes that were significantly upregulated in aortic media from 8-week-old mice of

both progeria models (Fig. 33A). This analysis confirmed progerin-induced upregulation of Calr,

Ddit3, Dnajb9, Hspa5, Hsp90b1, and Pdia4 in VSMC-rich aortic media in both models (Fig. 33B, C).

Figure 33. Endoplasmic reticulum (ER) stress and unfolded protein response (UPR) activation in progerin-

expressing aortas from Apoe-/-

LmnaG609G/G609G

mice (ubiquitous progerin) and Apoe-/-

LmnaLCS/LCS

SM22αCre mice

(vascular smooth muscle cell (VSMC)-specific progerin). (A) Six ER stress/UPR pathway genes selected for qPCR

validation from among of those detected as differentially expressed in RNAseq in both models. (B, C) qPCR results in

pooled medial aortas (n=4) obtained from 8-week-old Apoe-/-

LmnaG609G/G609G

mice (B) and Apoe-/-

LmnaLCS/LCS

SM22αCre

mice (C) and their corresponding controls. Hprt and Gusb were used for normalization. Data are mean ± SEM. Statistical

differences were analyzed by one-tailed t-test. *P<0.05, **P<0.01, ***P<0.001.

We next assessed whether progerin activates the ER stress response and the UPR in other

organs. Consistent with the ubiquity of progerin expression in Apoe-/-LmnaG609G/G609G mice, induction

of ER stress response and the UPR was noted in some organs of these animals, with kidney being the

organ most affected and liver the least (Fig. 34A). As anticipated, no activation of this stress pathway

was detected in kidney, liver, spleen, or heart from Apoe-/-LmnaLCS/LCSSM22Cre mice, confirming the

specificity of the model (Fig. 34B). The variability in the ER stress and UPR activation across the

organs of Apoe-/-LmnaG609G/G609G mice may be in part attributed to different lamin A (and thus progerin)

expression levels associated with tissue stiffness (63, 64). Moreover, distinct organs and tissues may

Page 82: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

77

have different thresholds for tolerating misfolded protein load and therefore be more prone or resistant

to ER stress.

Figure 34. Endoplasmic reticulum (ER) stress and unfolded protein response (UPR) activation in different organs

from Apoe-/-

LmnaG609G/G609G

mice with ubiquitous progerin expression. qPCR results in organs from 8-week-old Apoe-/-

LmnaG609G/G609G

mice (A) and Apoe-/-

LmnaLCS/LCS

SM22αCre mice (B) and their corresponding controls (n=4 per condition).

Activation of some ER stress and UPR genes was observed in kidney, heart, and spleen of Apoe-/-

LmnaG609G/G609G

mice but

not in the same organs of Apoe-/-

LmnaLCS/LCS

SM22αCre mice. Hprt and Gusb were used for normalization. Data are mean ±

SEM. Statistical differences were analyzed by two-tailed t-test. *P<0.05, **P<0.01, ***P<0.001.

Previous in vitro and in vivo studies have identified numerous pathways potentially contributing

to HGPS. Most of them, however, are mutually dependent, making it challenging to distinguish primary

from secondary mechanisms. Our RNAseq analysis of pre-disease aortic media from the ubiquitous

and VSMC-specific progeria models identified the ER stress response and the related UPR as potential

driver mechanisms of atherosclerosis in progeria. We hypothesize that accumulation of misfolded

progerin due to defective post-translational processing triggers ER stress in VSMCs, which might be

exaggerated by medial deposits of lipids, also known to activate ER stress (165, 166). At early stages,

VSMCs attempt to restore homeostasis by activating the UPR, but cell death occurs when stress cannot

be resolved. Because of continuous crosstalk between stress pathways, ER stress can lead to activation

of other stress responses. For instance, ER stress can sensitize cells to DNA damage-induced apoptosis

(167), providing a possible link between our findings and those of previous studies showing increased

Page 83: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

78

DNA damage in HGPS cells (101). In accordance with recent studies in HGPS fibroblasts, which

identified the NRF2 pathway as a major progerin target (168), our RNAseq analysis detected NRF2-

mediated oxidative stress response as one of the main pathways affected by progerin expression in

VSMCs. Oxidative stress induced by NRF2 is connected to ER stress through the protein kinase PERK,

which phosphorylates the transcription factor NRF2 in response to ER stress (169). Moreover, ER

stress is associated with inflammation, autophagy and mitochondrial dysfunction (170), processes that

are affected in progeria (171-173). Our results identify an important upstream mechanism in VSMCs,

which may link various stress pathways previously described in progeria. In vitro studies are thus

warranted to further explore in detail the mechanisms linking ER stress, UPR and VSMC death in the

setting of progeria, and the role of oxidized LDLs in this process. These studies could be performed

with primary VSMCs from wild-type and progerin-expressing mice, and VSMCs differentiated from

control and HGPS iPSCs (available from The Progeria Research Foundation).

IV.9. Therapeutic effects of ER stress response targeting in progeroid mice

Our RNAseq results strongly suggested a key role of ER stress response and the UPR as drivers

of progerin-induced VSMC death and enhanced atherosclerosis in the ubiquitous and VSMC-specific

progeria models. We therefore examined the potential benefits of targeting this pathway by chemical

chaperone treatment. We selected TUDCA, a bile acid previously proven to efficiently alleviate ER

stress and ameliorate experimental diabetes (174), aortic valve calcification (175), and myocardial

infarction (176).

Before using progeric mice, we conducted a pilot study with Apoe-/-Lmna+/+ control mice fed

normal chow to monitor for any adverse effects of prolonged TUDCA treatment. Animals received

TUDCA (400 mg/kg) or PBS intraperitoneal injections 3 times weekly starting at 6 weeks of age.

TUDCA- and PBS-treated mice appeared healthy and did not show any overt pathology. However,

weight gain in the TUDCA-treated group was slightly diminished (Fig. 35A), likely due to an increase

in energy expenditure, as described previously by da-Silva et al. (177). Analysis at 26 weeks of age

revealed similar blood pressure in both experimental groups (Fig. 35B), and the absence of blood

pressure and heart rate instability and variability (Fig. 35C). Similarly, except for a borderline

significant reduction in the PR segment in TUDCA-treated mice, all ECG parameters at 26 weeks of

age were similar in both experimental groups (Fig. 35D).

Page 84: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

79

Figure 35. Prolonged tauroursodeoxycholic acid (TUDCA) treatment of Apoe-/-Lmna+/+ mice does not cause any

severe alteration in blood pressure or electrocardiogram (ECG) parameters. Apoe-/-Lmna+/+ mice fed normal chow

received either TUDCA (400 mg/kg) or PBS intraperitoneal injections 3 times a week starting at 6 weeks of age. (A) Body

weight curves for TUDCA-treated and untreated mice. P<0.0001 (B) Heart rate, systolic and diastolic blood pressure

analysis for TUDCA-treated and untreated mice at 26 weeks of age. (C) Variability in the heart rate, systolic and diastolic

blood pressure for 26-week-old TUDCA-treated and untreated mice measured as coefficient of variation (CV). (D) ECG

parameters for 26-week-old TUDCA-treated and untreated mice. n=5. Data are presented as mean ± SEM. Statistical

analysis was performed by two-tailed t-test.

Necropsies of 27-week-old TUDCA- and PBS-treated mice did not reveal pathology in the

intraperitoneal cavity (Fig. 36A), which might arise because of the multiple injections or the drug per

se. We next analyzed the effect of TUDCA on atherosclerosis development as Erbay et al. found that

the chemical chaperone 4-phenyl butyric acid (PBA) can delay atherosclerotic plaque formation via

alleviating ER stress in macrophages (178). Importantly, this effect of PBA on atherosclerosis was

independent of changes in lipids, lipoprotein profiles, glucose and insulin levels in the circulation (178).

In agreement with these findings, we found a weak tendency toward reduced atherosclerosis in the

aortic arch in mice treated with TUDCA, although differences did not reach statistical significance

Page 85: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

80

(Fig. 36B). Our pilot study also revealed similar fasting serum levels of total and free cholesterol in

both experimental groups, and significantly lower LDL in TUDCA-treated animals (Fig. 36C).

Figure 36. Prolonged tauroursodeoxycholic acid (TUDCA) treatment of Apoe-/-Lmna+/+ mice does not significantly

affect atherosclerosis and has only minor effect on the lipid profile. Apoe-/-Lmna+/+ mice fed normal chow received

either TUDCA (400 mg/kg) or PBS intraperitoneal injections 3 times a week starting at 6 weeks of age. Mice were sacrificed

at 27 weeks of age. (A) Representative photographs of intraperitoneal cavity of TUDCA-treated and untreated mice. (B)

Representative images of Oil Red O-stained aortas of TUDCA-treated and untreated mice; graphs show quantification of

atherosclerosis burden in aortic arch and thoracic aorta. (C) Fasting serum levels of total cholesterol, free cholesterol, low-

density lipoprotein (LDL), and high-density lipoprotein (HDL) in 27-week-old TUDCA-treated and mice. n=5. Data in B

are shown as median; data in C are mean ± SEM. Statistical differences were analyzed by two-tailed Mann-Whitney test in

B and two-tailed t-test in C. *P<0.05.

We also observed that TUDCA induced some minor changes in the hematological parameters,

including higher lymphocyte and lower monocyte percentage (Fig. 37). Whether these alterations are

a cause or a consequence of diminished atherosclerosis burden should be addressed in a future study.

Page 86: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

81

Figure 37. Prolonged tauroursodeoxycholic acid (TUDCA) treatment of Apoe-/-Lmna+/+ mice does not cause any

significant alterations in hematological parameters. Apoe-/-Lmna+/+ mice fed normal chow received either TUDCA (400

mg/kg) or PBS intraperitoneal injections 3 times a week starting at 6 weeks of age. Hematology (fasting) results for 27-

week-old TUDCA-treated and untreated (PBS) mice; n=5. Data are shown as median. Statistical differences were analyzed

by two-tailed Mann-Whitney test.

After confirming that prolonged TUDCA treatment did not trigger any deleterious side effects,

we performed experiments with ubiquitous and VSMC-specific progeroid mouse models. Animals

were fed HFD for 8 weeks starting at 8 weeks of age. Additionally, mice received intraperitoneal

injections of TUDCA (400 mg/kg) 3 times weekly starting at 6 weeks of age in the case of Apoe-/-

LmnaG609G/G609G and Apoe-/-Lmna+/+ or at 8 weeks of age in the case of Apoe-/-LmnaLCS/LCSSM22Cre

and Apoe-/-LmnaLCS/LCS mice. Control mice received PBS injections. TUDCA treatment in both HFD-

fed Apoe-/-LmnaG609G/G609G and Apoe-/-LmnaLCS/LCSSM22Cre mice inhibited atheroma lesion formation

in the thoracic aorta (Fig. 38A, B) and alleviated aortic VSMC loss and adventitial thickening (Fig.

38C-F).

Page 87: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

82

Figure 38. Tauroursodeoxycholic acid (TUDCA) treatment alleviates vascular pathology in progeroid mouse models.

Mice were treated with TUDCA or vehicle (PBS), starting at 6 weeks of age for Apoe-/-

Lmna+/+

and Apoe-/-

LmnaG609G/G609G

mice, and at 8 weeks of age for Apoe-/-

LmnaLCS/LCS

and Apoe-/-

LmnaLCS/LCS

SM22αCre mice. All mice were fed high-fat diet

for 8 weeks starting at 8 weeks of age. (A, B) Representative images of thoracic aortas stained with Oil Red O and

quantification of atherosclerosis burden in TUDCA-treated and untreated mice of the indicated genotypes (n=7-8 in A; n=6-

8 in B). (C, D) Representative immunofluorescence images of aortas stained with anti-smooth muscle actin (Sma) antibody

(red) and Hoechst3442 (blue). Graphs show quantification of vascular smooth muscle cell (VSMC) content in the media as

either % of Sma-positive area (top) or number of nuclei/mm2 (bottom); n=6 in C (Apoe-/-

LmnaG609G/G609G

), and n=4-5 in D

(Apoe-/-

LmnaLCS/LCS

SM22aCre). Scale bar, 50 µm. (E, F) Representative histology sections of hematoxylin & eosin (H&E)-

stained aortas. Graphs show quantification of adventitia-to-media thickness ratio; n=6 in E (Apoe-/-

LmnaG609G/G609G

), and

n=4-5 in F (Apoe-/-

LmnaLCS/LCS

SM22aCre). Scale bar, 100 µm. Data in A, B, E and F are shown as median with interquartile

range and minima and maxima; data in C and D are mean ± SEM. Statistical differences were analyzed by one-way ANOVA

with Tukey´s post hoc test in A and B, one-tailed t-test in C and D, and one-tailed Mann-Whitney test in E and F. *P<0.05,

**P<0.01, ***P<0.001. m: media, a: adventitia.

We next examined the effect of TUDCA on survival. Notably, TUDCA prolonged the median

lifespan of Apoe-/-LmnaLCS/LCSSM22Cre mice by 35% (median survival: 61.4 weeks in TUDCA-

Page 88: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

83

treated versus 45.35 weeks in untreated mice, P=0.0148), without affecting the survival of Apoe-/-

LmnaG609G/G609G mice (Fig. 39A).

Figure 39. Tauroursodeoxycholic acid (TUDCA) treatment extends lifespan in Apoe-/-

LmnaLCS/LCS

SM22αCre mice.

Mice were treated with TUDCA or vehicle (PBS), starting at 6 weeks of age for Apoe-/-

Lmna+/+

and Apoe-/-

LmnaG609G/G609G

mice, and at 8 weeks of age for Apoe-/-

LmnaLCS/LCS

and Apoe

-/-Lmna

LCS/LCSSM22αCre mice. Graphs show Kaplan-Meier

survival (A) and body weight (B) curves of TUDCA-treated and untreated (PBS) mice of the indicated genotypes (n=7-8

Apoe-/-

LmnaG609G/G609G

mice; n=11-12 Apoe-/-

LmnaLCS/LCS

SM22αCre mice); P=0.0148 for TUDCA-treated vs untreated

Apoe-/-

LmnaLCS/LCS

SM22αCre mice (median survival: 61.4 vs 45.35 weeks, respectively). Data in B are mean ± SEM.

Statistical differences were analyzed by log-rank test in A.

These results show that whereas TUDCA treatment ameliorated vasculopathy in both progeria

models, it failed to prolong lifespan in mice ubiquitously expressing progerin. In accord with this

finding, ePPi treatment was shown to prevent vascular calcification in LmnaG609G/G609G mice, but had

no effect on survival (60). This result may be explained by our previous studies on the cause of death

of progeric mice, showing that Apoe-/-LmnaLCS/LCSSM22Cre mice, unlike Apoe-/-LmnaG609G/G609G mice,

Page 89: ATHEROSCLEROSIS IN PROGERIA

IV. RESULTS AND DISCUSSION

84

die of atherosclerosis-related causes. Thus, prevention of atherosclerosis by TUDCA in Apoe-/-

LmnaLCS/LCSSM22Cre mice was also associated with a significant prolongation of lifespan.

Although we did not detect any evident adverse effects of TUDCA treatment, we did observe

that weight gain was lower in TUDCA-treated groups than in PBS-injected groups (Fig. 35A and Fig.

39B). Even though this diminished weight gain was usually not significant, it was present across

different experiments in all analyzed genotypes and was independent of diet. This observation is in

agreement with the work of da-Silva et al., who showed that some chemical chaperones, including

TUDCA, accelerate thyroid hormone activation and energy expenditure (177). Remarkably, progerin

can also increase energy expenditure and mitochondrial activity, contributing to lipodystrophy; thus,

caution should be exercised when calculating the TUDCA dose for progeric mice, to avoid possible

harmful effects on adipose tissue. Nevertheless, four weeks of oral TUDCA treatment in obese patients

was shown not to alter body weight and total body fat, indicating that the doses used in humans (1750

mg/day/person in this particular study) are unlikely to affect body weight and adipose tissue

homeostasis (179).

The chemical chaperone TUDCA is a water-soluble bile acid that has been used successfully in

Europe to treat cholestatic liver disease (180, 181). TUDCA represents around 0.13% of the bile acid

pool in human serum (182), and is a high-abundance bile acid in American and Asiatic black bears

(183). Various studies reported no adverse effects of TUDCA treatment (daily doses from 500 to 1750

mg per person) in obese, liver-transplanted, and primary biliary cirrhosis patients (179, 184-186). In

general, chemical chaperones show exceptional in vivo safety, and some of them have been approved

for clinical use by the U.S. Food and Drug Administration, for example, PBA for urea cycle disorders

(187, 188) and ursodeoxycholic acid or primary biliary cirrhosis (189-191). Importantly, chemical

chaperones have been used successfully in children (187, 188). Thus, the potential of chemical

chaperone treatment to ameliorate atherosclerosis and associated ischemic events should be explored

in children with HGPS. The use of a combination of drugs targeting different pathways may be an

effective, therapeutic strategy until approaches directly targeting progerin production become available

in humans.

Page 90: ATHEROSCLEROSIS IN PROGERIA

87

V. CONCLUSIONS

Page 91: ATHEROSCLEROSIS IN PROGERIA

V. CONCLUSIONS

89

In this PhD thesis, I have generated new mouse models to study the role of progerin in accelerating

atherosclerosis and premature death, and to identify the underlying mechanisms and develop new

therapies for the treatment of progeria in mice. The main conclusions derived from this thesis are the

following:

1. Apoe-/- LmnaG609G/G609G mice with ubiquitous progerin expression display features of premature

aging (shortened life span, reduced body weight, etc.), and exhibit early onset of vascular

disease (including enhanced atherosclerosis, VSMC loss, increased lipid and collagen content

in the media, and adventitial thickening). As these alterations mimic the main symptoms of

HGPS, these mice constitute the first available preclinical model to study atherosclerosis in the

context of HGPS.

2. Apoe-/- LmnaLCS/LCS SM22αCre mice with VSMC-specific progerin expression do not exhibit

premature aging, but present the same vasculopathies as those observed in the ubiquitous

progeria model (including accelerated atherosclerosis, VSMC loss, lipid retention in the media,

adventitial thickening, etc.), and shortened life span. These findings demonstrate that progerin

effects on VSMCs play a crucial role in CVD progression and premature death in HGPS.

3. Apoe-/- LmnaLCS/LCS LysMCre mice with macrophage (myeloid)-specific progerin expression do

not show any premature aging or vascular phenotype.

4. Atherosclerotic lesions in Apoe-/- LmnaG609G/G609G and Apoe-/- LmnaLCS/LCS SM22αCre mice

display characteristic features of vulnerable plaques, which may lead to myocardial infarction.

5. Apoe-/- LmnaLCS/LCS SM22αCre mice develop hypotension as they age.

6. Apoe-/- LmnaG609G/G609G mice, unlike Apoe-/- LmnaLCS/LCS SM22αCre mice, develop early cardiac

electrical defects (prolonged QRS complex, QT and QTc intervals) and arrhythmias, which can

lead to premature death.

7. Apoe-/- LmnaLCS/LCS SM22αCre mice, unlike Apoe-/- LmnaG609G/G609G mice, die of

atherosclerosis-related causes.

8. High-throughput RNAseq analysis identified ER stress response and the subsequent UPR as

major alterations in the tunica media of both Apoe-/- LmnaG609G/G609G and Apoe-/- LmnaLCS/LCS

SM22αCre mice.

9. Targeting ER stress response and the UPR with the chemical chaperone TUDCA inhibits

atherosclerosis, VSMC loss and adventitial thickening in Apoe-/- LmnaG609G/G609G and Apoe-/-

LmnaLCS/LCS SM22αCre mice, and extends lifespan in the VSMC-specific model by 35%. These

Page 92: ATHEROSCLEROSIS IN PROGERIA

V. CONCLUSIONS

90

findings establish the ER stress response and the UPR as driver mechanisms of progerin-

induced VSMC death and accelerated atherosclerosis.

In summary, our results with progeroid mouse models suggest the possibility of chemical

chaperone treatment for ameliorating atherosclerosis in children with HGPS. Since premature and

physiological aging share many pathologies (including CVD), and progerin has been detected at low

levels in cells and tissues of normally aging individuals, research on HGPS might shed light on the

mechanisms of normal aging.

Page 93: ATHEROSCLEROSIS IN PROGERIA

93

V. CONCLUSIONES

Page 94: ATHEROSCLEROSIS IN PROGERIA

V. CONCLUSIONES

95

En esta tesis doctoral hemos generado nuevos modelos de ratón para estudiar el papel de la progerina

en la aceleración de la aterosclerosis y la muerte prematura asociada a HGPS. Además, hemos

identificado los mecanismos subyacentes y hemos desarrollado nuevas terapias para tratar la

enfermedad en ratones progéricos. Las principales conclusiones derivadas de esta tesis son:

1. Los ratones Apoe-/- LmnaG609G/G609G con expresión ubicua de progerina muestran características

de envejecimiento prematuro (pérdida de peso corporal, menor longevidad, etc.) y desarrollan

con mucha rapidez enfermedad vascular (incluyendo aterosclerosis acelerada, pérdida de

CMLVs, aumento del contenido de colágeno y retención de lípidos en la capa media, y fibrosis

de la capa adventicia). Dado que estas alteraciones recapitulan los principales síntomas del

HGPS, estos ratones son el primer modelo preclínico disponible para estudiar la aterosclerosis

en el contexto de HGPS.

2. Los ratones Apoe-/-LmnaLCS/LCS SM22αCre con expresión de progerina específica en CMLVs no

muestran fenotipo progérico, pero presentan las mismas alteraciones vasculares observadas en

el modelo de expresión ubicua de progeria, incluyendo aterosclerosis acelerada, pérdida de

CMLVs, retención de lípidos en la capa media y fibrosis de la capa adventicia, así como menor

longevidad. Estos hallazgos demuestran que los efectos de la progerina en las CMLVs juegan

un papel crítico en la progresión de la ECV y la muerte prematura asociadas a HGPS.

3. Los ratones Apoe-/-LmnaLCS/LCS LysMCre con expresión de progerina específica en macrófagos

(línea mieloide) no muestran envejecimiento prematuro ni patología vascular.

4. Las lesiones ateroscleróticas en ratones Apoe-/- LmnaG609G/G609G y Apoe-/-LmnaLCS/LCS SM22αCre

presentan características de placas vulnerables, que pueden provocar infarto de miocardio.

5. Los ratones Apoe-/-LmnaLCS/LCS SM22αCre desarrollan hipotensión a medida que envejecen.

6. Los ratones Apoe-/- LmnaG609G/G609G, a diferencia de Apoe-/-LmnaLCS/LCS SM22αCre, desarrollan

tempranamente alteraciones electrocardiográficas (prolongación del complejo QRS, y de los

intervalos QT y QTc) y arritmias. Estas anomalías pueden provocar muerte prematura.

7. Los ratones Apoe-/-LmnaLCS/LCS SM22αCre, a diferencia de Apoe-/- LmnaG609G/G609G, mueren por

problemas relacionadas con la aterosclerosis.

8. Mediante RNAseq hemos identificado un aumento significativo de genes implicados en el

estrés de RE y la respuesta a proteínas desplegadas (UPR) en la túnica de media de ratones con

expresión de progerina ubicua (Apoe-/- LmnaG609G/G609G) y específica de CMLVs (Apoe-/-

LmnaLCS/LCS SM22αCre).

Page 95: ATHEROSCLEROSIS IN PROGERIA

V. CONCLUSIONES

96

9. El tratamiento con TUDCA, una chaperona química que mejora la supervivencia celular en

situaciones de estrés de RE y UPR, inhibe la aterosclerosis, la pérdida de CMLVs y el

engrosamiento de la adventicia en ratones Apoe-/- LmnaG609G/G609G y Apoe-/-LmnaLCS/LCS

SM22αCre. Además, TUDCA prolonga en un 35% la supervivencia de los ratones Apoe-/-

LmnaLCS/LCS SM22αCre con expresión de progerina específica en CMLVs. Estos resultados

identifican la respuesta al estrés de RE y la UPR como un mecanismo clave en la inducción de

muerte de las CMLVs y la aceleración de la aterosclerosis provocadas por progerina.

En resumen, nuestros resultados con modelos de ratón progérico sugieren la posibilidad de

tratamiento con chaperonas químicas para mejorar la aterosclerosis en niños con HGPS. Teniendo en

cuenta que el envejecimiento prematuro y fisiológico comparten muchos mecanismos y patologías

(incluyendo ECV), y que se han detectado niveles bajos de expresión de progerina en células y tejidos

de individuos sin HGPS, nuestra investigación en HGPS podría arrojar luz sobre el envejecimiento

normal.

Page 96: ATHEROSCLEROSIS IN PROGERIA

99

VI. REFERENCES

Page 97: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

101

1. E. G. Lakatta, D. Levy, Arterial and cardiac aging: major shareholders in cardiovascular

disease enterprises: Part I: aging arteries: a "set up" for vascular disease. Circulation 107,

139-146 (2003).

2. S. S. Lim, T. Vos, A. D. Flaxman, G. Danaei, K. Shibuya, H. Adair-Rohani, M. Amann, H. R.

Anderson, K. G. Andrews, M. Aryee, C. Atkinson, L. J. Bacchus, A. N. Bahalim, K.

Balakrishnan, J. Balmes, S. Barker-Collo, A. Baxter, M. L. Bell, J. D. Blore, F. Blyth, C.

Bonner, G. Borges, R. Bourne, M. Boussinesq, M. Brauer, P. Brooks, N. G. Bruce, B.

Brunekreef, C. Bryan-Hancock, C. Bucello, R. Buchbinder, F. Bull, R. T. Burnett, T. E.

Byers, B. Calabria, J. Carapetis, E. Carnahan, Z. Chafe, F. Charlson, H. Chen, J. S. Chen, A.

T. Cheng, J. C. Child, A. Cohen, K. E. Colson, B. C. Cowie, S. Darby, S. Darling, A. Davis,

L. Degenhardt, F. Dentener, D. C. Des Jarlais, K. Devries, M. Dherani, E. L. Ding, E. R.

Dorsey, T. Driscoll, K. Edmond, S. E. Ali, R. E. Engell, P. J. Erwin, S. Fahimi, G. Falder, F.

Farzadfar, A. Ferrari, M. M. Finucane, S. Flaxman, F. G. Fowkes, G. Freedman, M. K.

Freeman, E. Gakidou, S. Ghosh, E. Giovannucci, G. Gmel, K. Graham, R. Grainger, B. Grant,

D. Gunnell, H. R. Gutierrez, W. Hall, H. W. Hoek, A. Hogan, H. D. Hosgood, 3rd, D. Hoy, H.

Hu, B. J. Hubbell, S. J. Hutchings, S. E. Ibeanusi, G. L. Jacklyn, R. Jasrasaria, J. B. Jonas, H.

Kan, J. A. Kanis, N. Kassebaum, N. Kawakami, Y. H. Khang, S. Khatibzadeh, J. P. Khoo, C.

Kok, F. Laden, R. Lalloo, Q. Lan, T. Lathlean, J. L. Leasher, J. Leigh, Y. Li, J. K. Lin, S. E.

Lipshultz, S. London, R. Lozano, Y. Lu, J. Mak, R. Malekzadeh, L. Mallinger, W. Marcenes,

L. March, R. Marks, R. Martin, P. McGale, J. McGrath, S. Mehta, G. A. Mensah, T. R.

Merriman, R. Micha, C. Michaud, V. Mishra, K. Mohd Hanafiah, A. A. Mokdad, L.

Morawska, D. Mozaffarian, T. Murphy, M. Naghavi, B. Neal, P. K. Nelson, J. M. Nolla, R.

Norman, C. Olives, S. B. Omer, J. Orchard, R. Osborne, B. Ostro, A. Page, K. D. Pandey, C.

D. Parry, E. Passmore, J. Patra, N. Pearce, P. M. Pelizzari, M. Petzold, M. R. Phillips, D.

Pope, C. A. Pope, 3rd, J. Powles, M. Rao, H. Razavi, E. A. Rehfuess, J. T. Rehm, B. Ritz, F.

P. Rivara, T. Roberts, C. Robinson, J. A. Rodriguez-Portales, I. Romieu, R. Room, L. C.

Rosenfeld, A. Roy, L. Rushton, J. A. Salomon, U. Sampson, L. Sanchez-Riera, E. Sanman, A.

Sapkota, S. Seedat, P. Shi, K. Shield, R. Shivakoti, G. M. Singh, D. A. Sleet, E. Smith, K. R.

Smith, N. J. Stapelberg, K. Steenland, H. Stockl, L. J. Stovner, K. Straif, L. Straney, G. D.

Thurston, J. H. Tran, R. Van Dingenen, A. van Donkelaar, J. L. Veerman, L. Vijayakumar, R.

Weintraub, M. M. Weissman, R. A. White, H. Whiteford, S. T. Wiersma, J. D. Wilkinson, H.

C. Williams, W. Williams, N. Wilson, A. D. Woolf, P. Yip, J. M. Zielinski, A. D. Lopez, C. J.

Murray, M. Ezzati, M. A. AlMazroa, Z. A. Memish, A comparative risk assessment of burden

Page 98: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

102

of disease and injury attributable to 67 risk factors and risk factor clusters in 21 regions, 1990-

2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 380, 2224-

2260 (2012).

3. E. J. Benjamin, M. J. Blaha, S. E. Chiuve, M. Cushman, S. R. Das, R. Deo, S. D. de Ferranti,

J. Floyd, M. Fornage, C. Gillespie, C. R. Isasi, M. C. Jimenez, L. C. Jordan, S. E. Judd, D.

Lackland, J. H. Lichtman, L. Lisabeth, S. Liu, C. T. Longenecker, R. H. Mackey, K.

Matsushita, D. Mozaffarian, M. E. Mussolino, K. Nasir, R. W. Neumar, L. Palaniappan, D. K.

Pandey, R. R. Thiagarajan, M. J. Reeves, M. Ritchey, C. J. Rodriguez, G. A. Roth, W. D.

Rosamond, C. Sasson, A. Towfighi, C. W. Tsao, M. B. Turner, S. S. Virani, J. H. Voeks, J. Z.

Willey, J. T. Wilkins, J. H. Wu, H. M. Alger, S. S. Wong, P. Muntner, C. American Heart

Association Statistics, S. Stroke Statistics, Heart Disease and Stroke Statistics-2017 Update: A

Report From the American Heart Association. Circulation 135, e146-e603 (2017).

4. R. Virmani, A. P. Avolio, W. J. Mergner, M. Robinowitz, E. E. Herderick, J. F. Cornhill, S.

Y. Guo, T. H. Liu, D. Y. Ou, M. O'Rourke, Effect of aging on aortic morphology in

populations with high and low prevalence of hypertension and atherosclerosis. Comparison

between occidental and Chinese communities. The American journal of pathology 139, 1119-

1129 (1991).

5. M. A. Lim, R. R. Townsend, Arterial compliance in the elderly: its effect on blood pressure

measurement and cardiovascular outcomes. Clinics in geriatric medicine 25, 191-205 (2009).

6. C. Vlachopoulos, K. Aznaouridis, C. Stefanadis, Prediction of cardiovascular events and all-

cause mortality with arterial stiffness: a systematic review and meta-analysis. Journal of the

American College of Cardiology 55, 1318-1327 (2010).

7. S. J. Zieman, V. Melenovsky, D. A. Kass, Mechanisms, pathophysiology, and therapy of

arterial stiffness. Arteriosclerosis, thrombosis, and vascular biology 25, 932-943 (2005).

8. Z. Li, J. Froehlich, Z. S. Galis, E. G. Lakatta, Increased expression of matrix

metalloproteinase-2 in the thickened intima of aged rats. Hypertension 33, 116-123 (1999).

9. G. K. Hansson, P. Libby, I. Tabas, Inflammation and plaque vulnerability. Journal of internal

medicine 278, 483-493 (2015).

10. G. P. Kwon, J. L. Schroeder, M. J. Amar, A. T. Remaley, R. S. Balaban, Contribution of

macromolecular structure to the retention of low-density lipoprotein at arterial branch points.

Circulation 117, 2919-2927 (2008).

Page 99: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

103

11. D. R. Greaves, S. Gordon, Thematic review series: the immune system and atherogenesis.

Recent insights into the biology of macrophage scavenger receptors. Journal of lipid research

46, 11-20 (2005).

12. L. Hegyi, J. N. Skepper, N. R. Cary, M. J. Mitchinson, Foam cell apoptosis and the

development of the lipid core of human atherosclerosis. The Journal of pathology 180, 423-

429 (1996).

13. C. Stefanadis, C. K. Antoniou, D. Tsiachris, P. Pietri, Coronary Atherosclerotic Vulnerable

Plaque: Current Perspectives. Journal of the American Heart Association 6, (2017).

14. J. Hutchinson, Congenital Absence of Hair and Mammary Glands with Atrophic Condition of

the Skin and its Appendages, in a Boy whose Mother had been almost wholly Bald from

Alopecia Areata from the age of Six. Medico-chirurgical transactions 69, 473-477 (1886).

15. H. Gilford, On a Condition of Mixed Premature and Immature Development. Medico-

chirurgical transactions 80, 17-46 25 (1897).

16. R. C. Hennekam, Hutchinson-Gilford progeria syndrome: review of the phenotype. American

journal of medical genetics. Part A 140, 2603-2624 (2006).

17. M. A. Merideth, L. B. Gordon, S. Clauss, V. Sachdev, A. C. Smith, M. B. Perry, C. C.

Brewer, C. Zalewski, H. J. Kim, B. Solomon, B. P. Brooks, L. H. Gerber, M. L. Turner, D. L.

Domingo, T. C. Hart, J. Graf, J. C. Reynolds, A. Gropman, J. A. Yanovski, M. Gerhard-

Herman, F. S. Collins, E. G. Nabel, R. O. Cannon, 3rd, W. A. Gahl, W. J. Introne, Phenotype

and course of Hutchinson-Gilford progeria syndrome. The New England journal of medicine

358, 592-604 (2008).

18. N. J. Ullrich, L. B. Gordon, Hutchinson-Gilford progeria syndrome. Handb Clin Neurol 132,

249-264 (2015).

19. L. B. Gordon, I. A. Harten, M. E. Patti, A. H. Lichtenstein, Reduced adiponectin and HDL

cholesterol without elevated C-reactive protein: clues to the biology of premature

atherosclerosis in Hutchinson-Gilford Progeria Syndrome. The Journal of pediatrics 146, 336-

341 (2005).

20. M. Olive, I. Harten, R. Mitchell, J. K. Beers, K. Djabali, K. Cao, M. R. Erdos, C. Blair, B.

Funke, L. Smoot, M. Gerhard-Herman, J. T. Machan, R. Kutys, R. Virmani, F. S. Collins, T.

N. Wight, E. G. Nabel, L. B. Gordon, Cardiovascular pathology in Hutchinson-Gilford

progeria: correlation with the vascular pathology of aging. Arteriosclerosis, thrombosis, and

vascular biology 30, 2301-2309 (2010).

Page 100: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

104

21. W. E. Stehbens, S. J. Wakefield, E. Gilbert-Barness, R. E. Olson, J. Ackerman, Histological

and ultrastructural features of atherosclerosis in progeria. Cardiovascular pathology : the

official journal of the Society for Cardiovascular Pathology 8, 29-39 (1999).

22. W. E. Stehbens, B. Delahunt, T. Shozawa, E. Gilbert-Barness, Smooth muscle cell depletion

and collagen types in progeric arteries. Cardiovascular pathology : the official journal of the

Society for Cardiovascular Pathology 10, 133-136 (2001).

23. M. Gerhard-Herman, L. B. Smoot, N. Wake, M. W. Kieran, M. E. Kleinman, D. T. Miller, A.

Schwartzman, A. Giobbie-Hurder, D. Neuberg, L. B. Gordon, Mechanisms of premature

vascular aging in children with Hutchinson-Gilford progeria syndrome. Hypertension 59, 92-

97 (2012).

24. J. Blacher, J. A. Staessen, X. Girerd, J. Gasowski, L. Thijs, L. Liu, J. G. Wang, R. H. Fagard,

M. E. Safar, Pulse pressure not mean pressure determines cardiovascular risk in older

hypertensive patients. Archives of internal medicine 160, 1085-1089 (2000).

25. A. Benetos, F. Thomas, L. Joly, J. Blacher, B. Pannier, C. Labat, P. Salvi, H. Smulyan, M. E.

Safar, Pulse pressure amplification a mechanical biomarker of cardiovascular risk. Journal of

the American College of Cardiology 55, 1032-1037 (2010).

26. P. M. Rothwell, Limitations of the usual blood-pressure hypothesis and importance of

variability, instability, and episodic hypertension. Lancet 375, 938-948 (2010).

27. P. M. Rothwell, S. C. Howard, E. Dolan, E. O'Brien, J. E. Dobson, B. Dahlof, P. S. Sever, N.

R. Poulter, Prognostic significance of visit-to-visit variability, maximum systolic blood

pressure, and episodic hypertension. Lancet 375, 895-905 (2010).

28. V. M. Silvera, L. B. Gordon, D. B. Orbach, S. E. Campbell, J. T. Machan, N. J. Ullrich,

Imaging characteristics of cerebrovascular arteriopathy and stroke in Hutchinson-Gilford

progeria syndrome. AJNR. American journal of neuroradiology 34, 1091-1097 (2013).

29. M. W. Kieran, L. Gordon, M. Kleinman, New approaches to progeria. Pediatrics 120, 834-

841 (2007).

30. J. Rivera-Torres, C. J. Calvo, A. Llach, G. Guzman-Martinez, R. Caballero, C. Gonzalez-

Gomez, L. J. Jimenez-Borreguero, J. A. Guadix, F. G. Osorio, C. Lopez-Otin, A. Herraiz-

Martinez, N. Cabello, A. Vallmitjana, R. Benitez, L. B. Gordon, J. Jalife, J. M. Perez-

Pomares, J. Tamargo, E. Delpon, L. Hove-Madsen, D. Filgueiras-Rama, V. Andres, Cardiac

electrical defects in progeroid mice and Hutchinson-Gilford progeria syndrome patients with

nuclear lamina alterations. Proc Natl Acad Sci U S A 113, E7250-E7259 (2016).

Page 101: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

105

31. K. Nair, P. Ramachandran, K. M. Krishnamoorthy, S. Dora, T. J. Achuthan, Hutchinson-

Gilford progeria syndrome with severe calcific aortic valve stenosis and calcific mitral valve.

J Heart Valve Dis 13, 866-869 (2004).

32. N. B. Hanumanthappa, G. Madhusudan, J. Mahimarangaiah, C. N. Manjunath, Hutchinson-

Gilford progeria syndrome with severe calcific aortic valve stenosis. Ann Pediatr Cardiol 4,

204-206 (2011).

33. M. Eriksson, W. T. Brown, L. B. Gordon, M. W. Glynn, J. Singer, L. Scott, M. R. Erdos, C.

M. Robbins, T. Y. Moses, P. Berglund, A. Dutra, E. Pak, S. Durkin, A. B. Csoka, M.

Boehnke, T. W. Glover, F. S. Collins, Recurrent de novo point mutations in lamin A cause

Hutchinson-Gilford progeria syndrome. Nature 423, 293-298 (2003).

34. A. De Sandre-Giovannoli, R. Bernard, P. Cau, C. Navarro, J. Amiel, I. Boccaccio, S. Lyonnet,

C. L. Stewart, A. Munnich, M. Le Merrer, N. Levy, Lamin a truncation in Hutchinson-Gilford

progeria. Science 300, 2055 (2003).

35. F. Lin, H. J. Worman, Structural organization of the human gene encoding nuclear lamin A

and nuclear lamin C. The Journal of biological chemistry 268, 16321-16326 (1993).

36. K. Furukawa, H. Inagaki, Y. Hotta, Identification and cloning of an mRNA coding for a germ

cell-specific A-type lamin in mice. Exp Cell Res 212, 426-430 (1994).

37. B. M. Machiels, A. H. Zorenc, J. M. Endert, H. J. Kuijpers, G. J. van Eys, F. C. Ramaekers, J.

L. Broers, An alternative splicing product of the lamin A/C gene lacks exon 10. The Journal

of biological chemistry 271, 9249-9253 (1996).

38. V. Andres, J. M. Gonzalez, Role of A-type lamins in signaling, transcription, and chromatin

organization. The Journal of cell biology 187, 945-957 (2009).

39. L. A. Beck, T. J. Hosick, M. Sinensky, Isoprenylation is required for the processing of the

lamin A precursor. The Journal of cell biology 110, 1489-1499 (1990).

40. M. Sinensky, K. Fantle, M. Trujillo, T. McLain, A. Kupfer, M. Dalton, The processing

pathway of prelamin A. Journal of cell science 107 ( Pt 1), 61-67 (1994).

41. D. Holtz, R. A. Tanaka, J. Hartwig, F. McKeon, The CaaX motif of lamin A functions in

conjunction with the nuclear localization signal to target assembly to the nuclear envelope.

Cell 59, 969-977 (1989).

42. D. P. Corrigan, D. Kuszczak, A. E. Rusinol, D. P. Thewke, C. A. Hrycyna, S. Michaelis, M.

S. Sinensky, Prelamin A endoproteolytic processing in vitro by recombinant Zmpste24. The

Biochemical journal 387, 129-138 (2005).

Page 102: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

106

43. A. M. Pendas, Z. Zhou, J. Cadinanos, J. M. Freije, J. Wang, K. Hultenby, A. Astudillo, A.

Wernerson, F. Rodriguez, K. Tryggvason, C. Lopez-Otin, Defective prelamin A processing

and muscular and adipocyte alterations in Zmpste24 metalloproteinase-deficient mice. Nat

Genet 31, 94-99 (2002).

44. K. Weber, U. Plessmann, P. Traub, Maturation of nuclear lamin A involves a specific

carboxy-terminal trimming, which removes the polyisoprenylation site from the precursor;

implications for the structure of the nuclear lamina. FEBS letters 257, 411-414 (1989).

45. S. Vidak, R. Foisner, Molecular insights into the premature aging disease progeria.

Histochemistry and cell biology 145, 401-417 (2016).

46. S. Gonzalo, R. Kreienkamp, P. Askjaer, Hutchinson-Gilford Progeria Syndrome: A premature

aging disease caused by LMNA gene mutations. Ageing research reviews 33, 18-29 (2017).

47. C. L. Navarro, A. De Sandre-Giovannoli, R. Bernard, I. Boccaccio, A. Boyer, D. Genevieve,

S. Hadj-Rabia, C. Gaudy-Marqueste, H. S. Smitt, P. Vabres, L. Faivre, A. Verloes, T. Van

Essen, E. Flori, R. Hennekam, F. A. Beemer, N. Laurent, M. Le Merrer, P. Cau, N. Levy,

Lamin A and ZMPSTE24 (FACE-1) defects cause nuclear disorganization and identify

restrictive dermopathy as a lethal neonatal laminopathy. Hum Mol Genet 13, 2493-2503

(2004).

48. C. L. Navarro, J. Cadinanos, A. De Sandre-Giovannoli, R. Bernard, S. Courrier, I. Boccaccio,

A. Boyer, W. J. Kleijer, A. Wagner, F. Giuliano, F. A. Beemer, J. M. Freije, P. Cau, R. C.

Hennekam, C. Lopez-Otin, C. Badens, N. Levy, Loss of ZMPSTE24 (FACE-1) causes

autosomal recessive restrictive dermopathy and accumulation of Lamin A precursors. Hum

Mol Genet 14, 1503-1513 (2005).

49. C. L. Moulson, G. Go, J. M. Gardner, A. C. van der Wal, J. H. Smitt, J. M. van Hagen, J. H.

Miner, Homozygous and compound heterozygous mutations in ZMPSTE24 cause the

laminopathy restrictive dermopathy. J Invest Dermatol 125, 913-919 (2005).

50. A. K. Agarwal, J. P. Fryns, R. J. Auchus, A. Garg, Zinc metalloproteinase, ZMPSTE24, is

mutated in mandibuloacral dysplasia. Hum Mol Genet 12, 1995-2001 (2003).

51. S. Reddy, L. Comai, Lamin A, farnesylation and aging. Exp Cell Res 318, 1-7 (2012).

52. C. Lefevre, M. Auclair, F. Boccara, J. P. Bastard, J. Capeau, C. Vigouroux, M. Caron-

Debarle, Premature senescence of vascular cells is induced by HIV protease inhibitors:

implication of prelamin A and reversion by statin. Arteriosclerosis, thrombosis, and vascular

biology 30, 2611-2620 (2010).

Page 103: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

107

53. P. Afonso, M. Auclair, F. Boccara, M. C. Vantyghem, C. Katlama, J. Capeau, C. Vigouroux,

M. Caron-Debarle, LMNA mutations resulting in lipodystrophy and HIV protease inhibitors

trigger vascular smooth muscle cell senescence and calcification: Role of ZMPSTE24

downregulation. Atherosclerosis 245, 200-211 (2016).

54. P. Scaffidi, T. Misteli, Lamin A-dependent nuclear defects in human aging. Science 312,

1059-1063 (2006).

55. D. McClintock, D. Ratner, M. Lokuge, D. M. Owens, L. B. Gordon, F. S. Collins, K. Djabali,

The mutant form of lamin A that causes Hutchinson-Gilford progeria is a biomarker of

cellular aging in human skin. PloS one 2, e1269 (2007).

56. S. H. Yang, M. O. Bergo, J. I. Toth, X. Qiao, Y. Hu, S. Sandoval, M. Meta, P. Bendale, M. H.

Gelb, S. G. Young, L. G. Fong, Blocking protein farnesyltransferase improves nuclear

blebbing in mouse fibroblasts with a targeted Hutchinson-Gilford progeria syndrome

mutation. Proc Natl Acad Sci U S A 102, 10291-10296 (2005).

57. S. H. Yang, M. Meta, X. Qiao, D. Frost, J. Bauch, C. Coffinier, S. Majumdar, M. O. Bergo, S.

G. Young, L. G. Fong, A farnesyltransferase inhibitor improves disease phenotypes in mice

with a Hutchinson-Gilford progeria syndrome mutation. J Clin Invest 116, 2115-2121 (2006).

58. R. Varga, M. Eriksson, M. R. Erdos, M. Olive, I. Harten, F. Kolodgie, B. C. Capell, J. Cheng,

D. Faddah, S. Perkins, H. Avallone, H. San, X. Qu, S. Ganesh, L. B. Gordon, R. Virmani, T.

N. Wight, E. G. Nabel, F. S. Collins, Progressive vascular smooth muscle cell defects in a

mouse model of Hutchinson-Gilford progeria syndrome. Proc Natl Acad Sci U S A 103, 3250-

3255 (2006).

59. F. G. Osorio, C. L. Navarro, J. Cadinanos, I. C. Lopez-Mejia, P. M. Quiros, C. Bartoli, J.

Rivera, J. Tazi, G. Guzman, I. Varela, D. Depetris, F. de Carlos, J. Cobo, V. Andres, A. De

Sandre-Giovannoli, J. M. Freije, N. Levy, C. Lopez-Otin, Splicing-directed therapy in a new

mouse model of human accelerated aging. Science translational medicine 3, 106ra107 (2011).

60. R. Villa-Bellosta, J. Rivera-Torres, F. G. Osorio, R. Acin-Perez, J. A. Enriquez, C. Lopez-

Otin, V. Andres, Defective extracellular pyrophosphate metabolism promotes vascular

calcification in a mouse model of Hutchinson-Gilford progeria syndrome that is ameliorated

on pyrophosphate treatment. Circulation 127, 2442-2451 (2013).

61. J. M. Lee, C. Nobumori, Y. Tu, C. Choi, S. H. Yang, H. J. Jung, T. A. Vickers, F. Rigo, C. F.

Bennett, S. G. Young, L. G. Fong, Modulation of LMNA splicing as a strategy to treat

prelamin A diseases. J Clin Invest 126, 1592-1602 (2016).

Page 104: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

108

62. M. O. Bergo, B. Gavino, J. Ross, W. K. Schmidt, C. Hong, L. V. Kendall, A. Mohr, M. Meta,

H. Genant, Y. Jiang, E. R. Wisner, N. Van Bruggen, R. A. Carano, S. Michaelis, S. M.

Griffey, S. G. Young, Zmpste24 deficiency in mice causes spontaneous bone fractures,

muscle weakness, and a prelamin A processing defect. Proc Natl Acad Sci U S A 99, 13049-

13054 (2002).

63. J. Swift, I. L. Ivanovska, A. Buxboim, T. Harada, P. C. Dingal, J. Pinter, J. D. Pajerowski, K.

R. Spinler, J. W. Shin, M. Tewari, F. Rehfeldt, D. W. Speicher, D. E. Discher, Nuclear lamin-

A scales with tissue stiffness and enhances matrix-directed differentiation. Science 341,

1240104 (2013).

64. J. Swift, D. E. Discher, The nuclear lamina is mechano-responsive to ECM elasticity in

mature tissue. Journal of cell science 127, 3005-3015 (2014).

65. C. L. Moulson, L. G. Fong, J. M. Gardner, E. A. Farber, G. Go, A. Passariello, D. K. Grange,

S. G. Young, J. H. Miner, Increased progerin expression associated with unusual LMNA

mutations causes severe progeroid syndromes. Hum Mutat 28, 882-889 (2007).

66. J. Reunert, R. Wentzell, M. Walter, S. Jakubiczka, M. Zenker, T. Brune, S. Rust, T.

Marquardt, Neonatal progeria: increased ratio of progerin to lamin A leads to progeria of the

newborn. Eur J Hum Genet 20, 933-937 (2012).

67. J. Denecke, T. Brune, T. Feldhaus, H. Robenek, C. Kranz, R. J. Auchus, A. K. Agarwal, T.

Marquardt, A homozygous ZMPSTE24 null mutation in combination with a heterozygous

mutation in the LMNA gene causes Hutchinson-Gilford progeria syndrome (HGPS): insights

into the pathophysiology of HGPS. Hum Mutat 27, 524-531 (2006).

68. L. G. Fong, J. K. Ng, M. Meta, N. Cote, S. H. Yang, C. L. Stewart, T. Sullivan, A. Burghardt,

S. Majumdar, K. Reue, M. O. Bergo, S. G. Young, Heterozygosity for Lmna deficiency

eliminates the progeria-like phenotypes in Zmpste24-deficient mice. Proc Natl Acad Sci U S

A 101, 18111-18116 (2004).

69. S. H. Yang, S. Y. Chang, S. Ren, Y. Wang, D. A. Andres, H. P. Spielmann, L. G. Fong, S. G.

Young, Absence of progeria-like disease phenotypes in knock-in mice expressing a non-

farnesylated version of progerin. Hum Mol Genet 20, 436-444 (2011).

70. B. S. Davies, R. H. Barnes, 2nd, Y. Tu, S. Ren, D. A. Andres, H. P. Spielmann, J.

Lammerding, Y. Wang, S. G. Young, L. G. Fong, An accumulation of non-farnesylated

prelamin A causes cardiomyopathy but not progeria. Hum Mol Genet 19, 2682-2694 (2010).

71. J. I. Toth, S. H. Yang, X. Qiao, A. P. Beigneux, M. H. Gelb, C. L. Moulson, J. H. Miner, S. G.

Young, L. G. Fong, Blocking protein farnesyltransferase improves nuclear shape in

Page 105: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

109

fibroblasts from humans with progeroid syndromes. Proc Natl Acad Sci U S A 102, 12873-

12878 (2005).

72. L. G. Fong, D. Frost, M. Meta, X. Qiao, S. H. Yang, C. Coffinier, S. G. Young, A protein

farnesyltransferase inhibitor ameliorates disease in a mouse model of progeria. Science 311,

1621-1623 (2006).

73. B. C. Capell, M. Olive, M. R. Erdos, K. Cao, D. A. Faddah, U. L. Tavarez, K. N. Conneely,

X. Qu, H. San, S. K. Ganesh, X. Chen, H. Avallone, F. D. Kolodgie, R. Virmani, E. G. Nabel,

F. S. Collins, A farnesyltransferase inhibitor prevents both the onset and late progression of

cardiovascular disease in a progeria mouse model. Proc Natl Acad Sci U S A 105, 15902-

15907 (2008).

74. I. Varela, S. Pereira, A. P. Ugalde, C. L. Navarro, M. F. Suarez, P. Cau, J. Cadinanos, F. G.

Osorio, N. Foray, J. Cobo, F. de Carlos, N. Levy, J. M. Freije, C. Lopez-Otin, Combined

treatment with statins and aminobisphosphonates extends longevity in a mouse model of

human premature aging. Nature medicine 14, 767-772 (2008).

75. L. B. Gordon, M. E. Kleinman, D. T. Miller, D. S. Neuberg, A. Giobbie-Hurder, M. Gerhard-

Herman, L. B. Smoot, C. M. Gordon, R. Cleveland, B. D. Snyder, B. Fligor, W. R. Bishop, P.

Statkevich, A. Regen, A. Sonis, S. Riley, C. Ploski, A. Correia, N. Quinn, N. J. Ullrich, A.

Nazarian, M. G. Liang, S. Y. Huh, A. Schwartzman, M. W. Kieran, Clinical trial of a

farnesyltransferase inhibitor in children with Hutchinson-Gilford progeria syndrome. Proc

Natl Acad Sci U S A 109, 16666-16671 (2012).

76. L. B. Gordon, J. Massaro, R. B. D'Agostino, Sr., S. E. Campbell, J. Brazier, W. T. Brown, M.

E. Kleinman, M. W. Kieran, C. Progeria Clinical Trials, Impact of farnesylation inhibitors on

survival in Hutchinson-Gilford progeria syndrome. Circulation 130, 27-34 (2014).

77. L. B. Gordon, M. E. Kleinman, J. Massaro, R. B. D'Agostino, Sr., H. Shappell, M. Gerhard-

Herman, L. B. Smoot, C. M. Gordon, R. H. Cleveland, A. Nazarian, B. D. Snyder, N. J.

Ullrich, V. M. Silvera, M. G. Liang, N. Quinn, D. T. Miller, S. Y. Huh, A. A. Dowton, K.

Littlefield, M. M. Greer, M. W. Kieran, Clinical Trial of the Protein Farnesylation Inhibitors

Lonafarnib, Pravastatin, and Zoledronic Acid in Children With Hutchinson-Gilford Progeria

Syndrome. Circulation 134, 114-125 (2016).

78. A. Kalinowski, P. N. Yaron, Z. Qin, S. Shenoy, M. J. Buehler, M. Losche, K. N. Dahl,

Interfacial binding and aggregation of lamin A tail domains associated with Hutchinson-

Gilford progeria syndrome. Biophys Chem 195, 43-48 (2014).

Page 106: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

110

79. Z. Qin, A. Kalinowski, K. N. Dahl, M. J. Buehler, Structure and stability of the lamin A tail

domain and HGPS mutant. J Struct Biol 175, 425-433 (2011).

80. R. D. Goldman, D. K. Shumaker, M. R. Erdos, M. Eriksson, A. E. Goldman, L. B. Gordon, Y.

Gruenbaum, S. Khuon, M. Mendez, R. Varga, F. S. Collins, Accumulation of mutant lamin A

causes progressive changes in nuclear architecture in Hutchinson-Gilford progeria syndrome.

Proc Natl Acad Sci U S A 101, 8963-8968 (2004).

81. K. N. Dahl, P. Scaffidi, M. F. Islam, A. G. Yodh, K. L. Wilson, T. Misteli, Distinct structural

and mechanical properties of the nuclear lamina in Hutchinson-Gilford progeria syndrome.

Proc Natl Acad Sci U S A 103, 10271-10276 (2006).

82. S. J. Lee, Y. S. Jung, M. H. Yoon, S. M. Kang, A. Y. Oh, J. H. Lee, S. Y. Jun, T. G. Woo, H.

Y. Chun, S. K. Kim, K. J. Chung, H. Y. Lee, K. Lee, G. Jin, M. K. Na, N. C. Ha, C. Barcena,

J. M. Freije, C. Lopez-Otin, G. Y. Song, B. J. Park, Interruption of progerin-lamin A/C

binding ameliorates Hutchinson-Gilford progeria syndrome phenotype. J Clin Invest 126,

3879-3893 (2016).

83. M. X. Ibrahim, V. I. Sayin, M. K. Akula, M. Liu, L. G. Fong, S. G. Young, M. O. Bergo,

Targeting isoprenylcysteine methylation ameliorates disease in a mouse model of progeria.

Science 340, 1330-1333 (2013).

84. K. Cao, J. J. Graziotto, C. D. Blair, J. R. Mazzulli, M. R. Erdos, D. Krainc, F. S. Collins,

Rapamycin reverses cellular phenotypes and enhances mutant protein clearance in

Hutchinson-Gilford progeria syndrome cells. Sci Transl Med 3, 89ra58 (2011).

85. J. J. Graziotto, K. Cao, F. S. Collins, D. Krainc, Rapamycin activates autophagy in

Hutchinson-Gilford progeria syndrome: implications for normal aging and age-dependent

neurodegenerative disorders. Autophagy 8, 147-151 (2012).

86. S. J. Lee, S. H. Park, Arterial ageing. Korean Circ J 43, 73-79 (2013).

87. A. Buxboim, J. Swift, J. Irianto, K. R. Spinler, P. C. Dingal, A. Athirasala, Y. R. Kao, S. Cho,

T. Harada, J. W. Shin, D. E. Discher, Matrix elasticity regulates lamin-A,C phosphorylation

and turnover with feedback to actomyosin. Current biology : CB 24, 1909-1917 (2014).

88. J. T. Philip, K. N. Dahl, Nuclear mechanotransduction: response of the lamina to extracellular

stress with implications in aging. J Biomech 41, 3164-3170 (2008).

89. E. A. Booth, S. T. Spagnol, T. A. Alcoser, K. N. Dahl, Nuclear stiffening and chromatin

softening with progerin expression leads to an attenuated nuclear response to force. Soft

Matter 11, 6412-6418 (2015).

Page 107: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

111

90. V. L. Verstraeten, J. Y. Ji, K. S. Cummings, R. T. Lee, J. Lammerding, Increased

mechanosensitivity and nuclear stiffness in Hutchinson-Gilford progeria cells: effects of

farnesyltransferase inhibitors. Aging Cell 7, 383-393 (2008).

91. M. Song, H. San, S. A. Anderson, R. O. Cannon, 3rd, D. Orlic, Shear stress-induced

mechanotransduction protein deregulation and vasculopathy in a mouse model of progeria.

Stem Cell Res Ther 5, 41 (2014).

92. J. A. Brassard, N. Fekete, A. Garnier, C. A. Hoesli, Hutchinson-Gilford progeria syndrome as

a model for vascular aging. Biogerontology 17, 129-145 (2016).

93. T. Katsumoto, A. Mitsushima, T. Kurimura, The role of the vimentin intermediate filaments

in rat 3Y1 cells elucidated by immunoelectron microscopy and computer-graphic

reconstruction. Biol Cell 68, 139-146 (1990).

94. G. H. Liu, B. Z. Barkho, S. Ruiz, D. Diep, J. Qu, S. L. Yang, A. D. Panopoulos, K. Suzuki, L.

Kurian, C. Walsh, J. Thompson, S. Boue, H. L. Fung, I. Sancho-Martinez, K. Zhang, J. Yates,

3rd, J. C. Izpisua Belmonte, Recapitulation of premature ageing with iPSCs from Hutchinson-

Gilford progeria syndrome. Nature 472, 221-225 (2011).

95. D. Kinoshita, A. Nagasawa, I. Shimizu, T. K. Ito, Y. Yoshida, M. Tsuchida, A. Iwama, T.

Hayano, T. Minamino, Progerin impairs vascular smooth muscle cell growth via the DNA

damage response pathway. Oncotarget, (2017).

96. H. Zhang, Z. M. Xiong, K. Cao, Mechanisms controlling the smooth muscle cell death in

progeria via down-regulation of poly(ADP-ribose) polymerase 1. Proc Natl Acad Sci U S A

111, E2261-2270 (2014).

97. C. D. Ragnauth, D. T. Warren, Y. Liu, R. McNair, T. Tajsic, N. Figg, R. Shroff, J. Skepper,

C. M. Shanahan, Prelamin A acts to accelerate smooth muscle cell senescence and is a novel

biomarker of human vascular aging. Circulation 121, 2200-2210 (2010).

98. Y. Liu, I. Drozdov, R. Shroff, L. E. Beltran, C. M. Shanahan, Prelamin A accelerates vascular

calcification via activation of the DNA damage response and senescence-associated secretory

phenotype in vascular smooth muscle cells. Circ Res 112, e99-109 (2013).

99. A. M. Cobb, D. Larrieu, D. T. Warren, Y. Liu, S. Srivastava, A. J. Smith, R. P. Bowater, S. P.

Jackson, C. M. Shanahan, Prelamin A impairs 53BP1 nuclear entry by mislocalizing NUP153

and disrupting the Ran gradient. Aging Cell, (2016).

100. B. Liu, J. Wang, K. M. Chan, W. M. Tjia, W. Deng, X. Guan, J. D. Huang, K. M. Li, P. Y.

Chau, D. J. Chen, D. Pei, A. M. Pendas, J. Cadinanos, C. Lopez-Otin, H. F. Tse, C.

Page 108: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

112

Hutchison, J. Chen, Y. Cao, K. S. Cheah, K. Tryggvason, Z. Zhou, Genomic instability in

laminopathy-based premature aging. Nature medicine 11, 780-785 (2005).

101. B. Liu, Z. Wang, S. Ghosh, Z. Zhou, Defective ATM-Kap-1-mediated chromatin remodeling

impairs DNA repair and accelerates senescence in progeria mouse model. Aging Cell 12, 316-

318 (2013).

102. B. Liu, Z. Wang, L. Zhang, S. Ghosh, H. Zheng, Z. Zhou, Depleting the methyltransferase

Suv39h1 improves DNA repair and extends lifespan in a progeria mouse model. Nat Commun

4, 1868 (2013).

103. G. A. Garinis, G. T. van der Horst, J. Vijg, J. H. Hoeijmakers, DNA damage and ageing: new-

age ideas for an age-old problem. Nature cell biology 10, 1241-1247 (2008).

104. M. Salamat, P. K. Dhar, D. L. Neagu, J. B. Lyon, Aortic calcification in a patient with

hutchinson-gilford progeria syndrome. Pediatr Cardiol 31, 925-926 (2010).

105. J. C. Kovacic, P. Moreno, E. G. Nabel, V. Hachinski, V. Fuster, Cellular senescence, vascular

disease, and aging: part 2 of a 2-part review: clinical vascular disease in the elderly.

Circulation 123, 1900-1910 (2011).

106. I. Quiros-Gonzalez, P. Roman-Garcia, C. Alonso-Montes, S. Barrio-Vazquez, N. Carrillo-

Lopez, M. Naves-Diaz, M. I. Mora, F. J. Corrales, F. J. Lopez-Hernandez, M. P. Ruiz-Torres,

J. B. Cannata-Andia, J. L. Fernandez-Martin, Lamin A is involved in the development of

vascular calcification induced by chronic kidney failure and phosphorus load. Bone 84, 160-

168 (2016).

107. P. Scaffidi, T. Misteli, Lamin A-dependent misregulation of adult stem cells associated with

accelerated ageing. Nature cell biology 10, 452-459 (2008).

108. S. G. Frangos, V. Gahtan, B. Sumpio, Localization of atherosclerosis: role of hemodynamics.

Arch Surg 134, 1142-1149 (1999).

109. L. Rouleau, J. Rossi, R. L. Leask, The response of human aortic endothelial cells in a stenotic

hemodynamic environment: effect of duration, magnitude, and spatial gradients in wall shear

stress. J Biomech Eng 132, 071015 (2010).

110. N. Bonello-Palot, S. Simoncini, S. Robert, P. Bourgeois, F. Sabatier, N. Levy, F. Dignat-

George, C. Badens, Prelamin A accumulation in endothelial cells induces premature

senescence and functional impairment. Atherosclerosis 237, 45-52 (2014).

111. T. B. Kirkwood, S. N. Austad, Why do we age? Nature 408, 233-238 (2000).

112. N. Kourtis, N. Tavernarakis, Cellular stress response pathways and ageing: intricate molecular

relationships. The EMBO journal 30, 2520-2531 (2011).

Page 109: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

113

113. D. Lindholm, H. Wootz, L. Korhonen, ER stress and neurodegenerative diseases. Cell death

and differentiation 13, 385-392 (2006).

114. H. Yoshida, ER stress and diseases. The FEBS journal 274, 630-658 (2007).

115. J. H. Lin, P. Walter, T. S. Yen, Endoplasmic reticulum stress in disease pathogenesis. Annual

review of pathology 3, 399-425 (2008).

116. M. Schroder, R. J. Kaufman, The mammalian unfolded protein response. Annual review of

biochemistry 74, 739-789 (2005).

117. D. Ron, P. Walter, Signal integration in the endoplasmic reticulum unfolded protein response.

Nature reviews. Molecular cell biology 8, 519-529 (2007).

118. P. Walter, D. Ron, The unfolded protein response: from stress pathway to homeostatic

regulation. Science 334, 1081-1086 (2011).

119. C. R. Prostko, M. A. Brostrom, C. O. Brostrom, Reversible phosphorylation of eukaryotic

initiation factor 2 alpha in response to endoplasmic reticular signaling. Molecular and cellular

biochemistry 127-128, 255-265 (1993).

120. R. J. Kaufman, Stress signaling from the lumen of the endoplasmic reticulum: coordination of

gene transcriptional and translational controls. Genes & development 13, 1211-1233 (1999).

121. R. Y. Hampton, ER stress response: getting the UPR hand on misfolded proteins. Current

biology : CB 10, R518-521 (2000).

122. C. Hetz, The unfolded protein response: controlling cell fate decisions under ER stress and

beyond. Nature reviews. Molecular cell biology 13, 89-102 (2012).

123. E. Szegezdi, S. E. Logue, A. M. Gorman, A. Samali, Mediators of endoplasmic reticulum

stress-induced apoptosis. EMBO reports 7, 880-885 (2006).

124. A. Bertolotti, Y. Zhang, L. M. Hendershot, H. P. Harding, D. Ron, Dynamic interaction of

BiP and ER stress transducers in the unfolded-protein response. Nature cell biology 2, 326-

332 (2000).

125. H. P. Harding, I. Novoa, Y. Zhang, H. Zeng, R. Wek, M. Schapira, D. Ron, Regulated

translation initiation controls stress-induced gene expression in mammalian cells. Molecular

cell 6, 1099-1108 (2000).

126. D. Han, A. G. Lerner, L. Vande Walle, J. P. Upton, W. Xu, A. Hagen, B. J. Backes, S. A.

Oakes, F. R. Papa, IRE1alpha kinase activation modes control alternate endoribonuclease

outputs to determine divergent cell fates. Cell 138, 562-575 (2009).

127. J. Hollien, J. S. Weissman, Decay of endoplasmic reticulum-localized mRNAs during the

unfolded protein response. Science 313, 104-107 (2006).

Page 110: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

114

128. J. Hollien, J. H. Lin, H. Li, N. Stevens, P. Walter, J. S. Weissman, Regulated Ire1-dependent

decay of messenger RNAs in mammalian cells. The Journal of cell biology 186, 323-331

(2009).

129. G. Kroemer, G. Marino, B. Levine, Autophagy and the integrated stress response. Molecular

cell 40, 280-293 (2010).

130. K. Haze, H. Yoshida, H. Yanagi, T. Yura, K. Mori, Mammalian transcription factor ATF6 is

synthesized as a transmembrane protein and activated by proteolysis in response to

endoplasmic reticulum stress. Molecular biology of the cell 10, 3787-3799 (1999).

131. J. Ye, R. B. Rawson, R. Komuro, X. Chen, U. P. Dave, R. Prywes, M. S. Brown, J. L.

Goldstein, ER stress induces cleavage of membrane-bound ATF6 by the same proteases that

process SREBPs. Molecular cell 6, 1355-1364 (2000).

132. K. Haze, T. Okada, H. Yoshida, H. Yanagi, T. Yura, M. Negishi, K. Mori, Identification of

the G13 (cAMP-response-element-binding protein-related protein) gene product related to

activating transcription factor 6 as a transcriptional activator of the mammalian unfolded

protein response. The Biochemical journal 355, 19-28 (2001).

133. K. Lee, W. Tirasophon, X. Shen, M. Michalak, R. Prywes, T. Okada, H. Yoshida, K. Mori, R.

J. Kaufman, IRE1-mediated unconventional mRNA splicing and S2P-mediated ATF6

cleavage merge to regulate XBP1 in signaling the unfolded protein response. Genes &

development 16, 452-466 (2002).

134. C. E. Shamu, P. Walter, Oligomerization and phosphorylation of the Ire1p kinase during

intracellular signaling from the endoplasmic reticulum to the nucleus. The EMBO journal 15,

3028-3039 (1996).

135. M. Calfon, H. Zeng, F. Urano, J. H. Till, S. R. Hubbard, H. P. Harding, S. G. Clark, D. Ron,

IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1

mRNA. Nature 415, 92-96 (2002).

136. H. Yoshida, T. Matsui, A. Yamamoto, T. Okada, K. Mori, XBP1 mRNA is induced by ATF6

and spliced by IRE1 in response to ER stress to produce a highly active transcription factor.

Cell 107, 881-891 (2001).

137. A. H. Lee, N. N. Iwakoshi, L. H. Glimcher, XBP-1 regulates a subset of endoplasmic

reticulum resident chaperone genes in the unfolded protein response. Molecular and cellular

biology 23, 7448-7459 (2003).

Page 111: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

115

138. D. Acosta-Alvear, Y. Zhou, A. Blais, M. Tsikitis, N. H. Lents, C. Arias, C. J. Lennon, Y.

Kluger, B. D. Dynlacht, XBP1 controls diverse cell type- and condition-specific

transcriptional regulatory networks. Molecular cell 27, 53-66 (2007).

139. R. Sriburi, S. Jackowski, K. Mori, J. W. Brewer, XBP1: a link between the unfolded protein

response, lipid biosynthesis, and biogenesis of the endoplasmic reticulum. The Journal of cell

biology 167, 35-41 (2004).

140. Z. Liu, Y. Lv, N. Zhao, G. Guan, J. Wang, Protein kinase R-like ER kinase and its role in

endoplasmic reticulum stress-decided cell fate. Cell death & disease 6, e1822 (2015).

141. N. Naidoo, ER and aging-Protein folding and the ER stress response. Ageing research reviews

8, 150-159 (2009).

142. K. Ameri, A. L. Harris, Activating transcription factor 4. The international journal of

biochemistry & cell biology 40, 14-21 (2008).

143. I. Novoa, H. Zeng, H. P. Harding, D. Ron, Feedback inhibition of the unfolded protein

response by GADD34-mediated dephosphorylation of eIF2alpha. The Journal of cell biology

153, 1011-1022 (2001).

144. N. Naidoo, The endoplasmic reticulum stress response and aging. Reviews in the

neurosciences 20, 23-37 (2009).

145. L. Wang, J. Meng, W. Cao, Q. Li, Y. Qiu, B. Sun, L. M. Li, Induction of apoptosis through

ER stress and TP53 in MCF-7 cells by the nanoparticle [Gd@C82(OH)22]n: A systems

biology study. Methods 67, 394-406 (2014).

146. J. Zhang, Q. Lian, G. Zhu, F. Zhou, L. Sui, C. Tan, R. A. Mutalif, R. Navasankari, Y. Zhang,

H. F. Tse, C. L. Stewart, A. Colman, A human iPSC model of Hutchinson Gilford Progeria

reveals vascular smooth muscle and mesenchymal stem cell defects. Cell Stem Cell 8, 31-45

(2011).

147. B. E. Clausen, C. Burkhardt, W. Reith, R. Renkawitz, I. Forster, Conditional gene targeting in

macrophages and granulocytes using LysMcre mice. Transgenic Res 8, 265-277 (1999).

148. R. Agarwal, Regulation of circadian blood pressure: from mice to astronauts. Current opinion

in nephrology and hypertension 19, 51-58 (2010).

149. F. Halberg, H. Mult, G. Cornelissen, D. Hillman, L. A. Beaty, S. Hong, O. Schwartzkopff, Y.

Watanabe, K. Otsuka, J. Siegelova, Chronobiologically Interpreted Ambulatory Blood

Pressure Monitoring in Health and Disease. Global advances in health and medicine 1, 66-

123 (2012).

150. S. Andrews, FastQC: a quality control tool for high throughput sequence data. 2010.

Page 112: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

116

151. M. Martin, Cutadapt removes adapter sequences from high-throughput sequencing reads.

2011 17, (2011).

152. B. Li, C. N. Dewey, RSEM: accurate transcript quantification from RNA-Seq data with or

without a reference genome. BMC Bioinformatics 12, 323 (2011).

153. M. D. Robinson, D. J. McCarthy, G. K. Smyth, edgeR: a Bioconductor package for

differential expression analysis of digital gene expression data. Bioinformatics 26, 139-140

(2010).

154. T. Hulsen, J. de Vlieg, W. Alkema, BioVenn - a web application for the comparison and

visualization of biological lists using area-proportional Venn diagrams. BMC Genomics 9,

488 (2008).

155. S. H. Yang, X. Qiao, E. Farber, S. Y. Chang, L. G. Fong, S. G. Young, Eliminating the

synthesis of mature lamin A reduces disease phenotypes in mice carrying a Hutchinson-

Gilford progeria syndrome allele. The Journal of biological chemistry 283, 7094-7099 (2008).

156. J. J. Fuster, A. I. Castillo, C. Zaragoza, B. Ibanez, V. Andres, Animal models of

atherosclerosis. Prog Mol Biol Transl Sci 105, 1-23 (2012).

157. P. Libby, G. K. Hansson, Inflammation and immunity in diseases of the arterial tree: players

and layers. Circ Res 116, 307-311 (2015).

158. I. C. Lopez-Mejia, M. de Toledo, C. Chavey, L. Lapasset, P. Cavelier, C. Lopez-Herrera, K.

Chebli, P. Fort, G. Beranger, L. Fajas, E. Z. Amri, F. Casas, J. Tazi, Antagonistic functions of

LMNA isoforms in energy expenditure and lifespan. EMBO reports 15, 529-539 (2014).

159. M. C. Clarke, N. Figg, J. J. Maguire, A. P. Davenport, M. Goddard, T. D. Littlewood, M. R.

Bennett, Apoptosis of vascular smooth muscle cells induces features of plaque vulnerability in

atherosclerosis. Nature medicine 12, 1075-1080 (2006).

160. S. A. Kamath, P. Meo Neto Jde, R. M. Canham, F. Uddin, K. H. Toto, L. L. Nelson, P. A.

Kaiser, J. A. de Lemos, M. H. Drazner, Low voltage on the electrocardiogram is a marker of

disease severity and a risk factor for adverse outcomes in patients with heart failure due to

systolic dysfunction. American heart journal 152, 355-361 (2006).

161. J. Gehrmann, S. Frantz, C. T. Maguire, M. Vargas, A. Ducharme, H. Wakimoto, R. T. Lee, C.

I. Berul, Electrophysiological characterization of murine myocardial ischemia and infarction.

Basic research in cardiology 96, 237-250 (2001).

162. N. Clausell, J. Butany, P. Gladstone, E. Lonn, P. Liu, C. Cardella, C. Feindel, P. A. Daly,

Myocardial vacuolization, a marker of ischemic injury, in surveillance cardiac biopsies

posttransplant: Correlations with morphologic vascular disease and endothelial dysfunction.

Page 113: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

117

Cardiovascular pathology : the official journal of the Society for Cardiovascular Pathology 5,

29-37 (1996).

163. R. B. Driesen, F. K. Verheyen, P. Dijkstra, F. Thone, J. P. Cleutjens, M. H. Lenders, F. C.

Ramaekers, M. Borgers, Structural remodelling of cardiomyocytes in the border zone of

infarcted rabbit heart. Molecular and cellular biochemistry 302, 225-232 (2007).

164. J. S. Pirolo, G. M. Hutchins, G. W. Moore, Myocyte vacuolization in infarct border zones is

reversible. The American journal of pathology 121, 444-450 (1985).

165. B. Feng, P. M. Yao, Y. Li, C. M. Devlin, D. Zhang, H. P. Harding, M. Sweeney, J. X. Rong,

G. Kuriakose, E. A. Fisher, A. R. Marks, D. Ron, I. Tabas, The endoplasmic reticulum is the

site of cholesterol-induced cytotoxicity in macrophages. Nature cell biology 5, 781-792

(2003).

166. D. A. Cunha, P. Hekerman, L. Ladriere, A. Bazarra-Castro, F. Ortis, M. C. Wakeham, F.

Moore, J. Rasschaert, A. K. Cardozo, E. Bellomo, L. Overbergh, C. Mathieu, R. Lupi, T. Hai,

A. Herchuelz, P. Marchetti, G. A. Rutter, D. L. Eizirik, M. Cnop, Initiation and execution of

lipotoxic ER stress in pancreatic beta-cells. Journal of cell science 121, 2308-2318 (2008).

167. C. Mlynarczyk, R. Fahraeus, Endoplasmic reticulum stress sensitizes cells to DNA damage-

induced apoptosis through p53-dependent suppression of p21(CDKN1A). Nat Commun 5,

5067 (2014).

168. N. Kubben, W. Zhang, L. Wang, T. C. Voss, J. Yang, J. Qu, G. H. Liu, T. Misteli, Repression

of the Antioxidant NRF2 Pathway in Premature Aging. Cell 165, 1361-1374 (2016).

169. S. B. Cullinan, D. Zhang, M. Hannink, E. Arvisais, R. J. Kaufman, J. A. Diehl, Nrf2 is a

direct PERK substrate and effector of PERK-dependent cell survival. Molecular and cellular

biology 23, 7198-7209 (2003).

170. D. Senft, Z. A. Ronai, UPR, autophagy, and mitochondria crosstalk underlies the ER stress

response. Trends Biochem Sci 40, 141-148 (2015).

171. F. G. Osorio, C. Barcena, C. Soria-Valles, A. J. Ramsay, F. de Carlos, J. Cobo, A. Fueyo, J.

M. Freije, C. Lopez-Otin, Nuclear lamina defects cause ATM-dependent NF-kappaB

activation and link accelerated aging to a systemic inflammatory response. Genes &

development 26, 2311-2324 (2012).

172. J. Rivera-Torres, R. Acin-Perez, P. Cabezas-Sanchez, F. G. Osorio, C. Gonzalez-Gomez, D.

Megias, C. Camara, C. Lopez-Otin, J. A. Enriquez, J. L. Luque-Garcia, V. Andres,

Identification of mitochondrial dysfunction in Hutchinson-Gilford progeria syndrome through

Page 114: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

118

use of stable isotope labeling with amino acids in cell culture. J Proteomics 91, 466-477

(2013).

173. G. Marino, A. P. Ugalde, N. Salvador-Montoliu, I. Varela, P. M. Quiros, J. Cadinanos, I. van

der Pluijm, J. M. Freije, C. Lopez-Otin, Premature aging in mice activates a systemic

metabolic response involving autophagy induction. Hum Mol Genet 17, 2196-2211 (2008).

174. U. Ozcan, E. Yilmaz, L. Ozcan, M. Furuhashi, E. Vaillancourt, R. O. Smith, C. Z. Gorgun, G.

S. Hotamisligil, Chemical chaperones reduce ER stress and restore glucose homeostasis in a

mouse model of type 2 diabetes. Science 313, 1137-1140 (2006).

175. Z. Cai, F. Li, W. Gong, W. Liu, Q. Duan, C. Chen, L. Ni, Y. Xia, K. Cianflone, N. Dong, D.

W. Wang, Endoplasmic reticulum stress participates in aortic valve calcification in

hypercholesterolemic animals. Arteriosclerosis, thrombosis, and vascular biology 33, 2345-

2354 (2013).

176. A. L. Rivard, C. J. Steer, B. T. Kren, C. M. Rodrigues, R. E. Castro, R. W. Bianco, W. C.

Low, Administration of tauroursodeoxycholic acid (TUDCA) reduces apoptosis following

myocardial infarction in rat. Am J Chin Med 35, 279-295 (2007).

177. W. S. da-Silva, S. Ribich, R. Arrojo e Drigo, M. Castillo, M. E. Patti, A. C. Bianco, The

chemical chaperones tauroursodeoxycholic and 4-phenylbutyric acid accelerate thyroid

hormone activation and energy expenditure. FEBS letters 585, 539-544 (2011).

178. E. Erbay, V. R. Babaev, J. R. Mayers, L. Makowski, K. N. Charles, M. E. Snitow, S. Fazio,

M. M. Wiest, S. M. Watkins, M. F. Linton, G. S. Hotamisligil, Reducing endoplasmic

reticulum stress through a macrophage lipid chaperone alleviates atherosclerosis. Nature

medicine 15, 1383-1391 (2009).

179. M. Kars, L. Yang, M. F. Gregor, B. S. Mohammed, T. A. Pietka, B. N. Finck, B. W.

Patterson, J. D. Horton, B. Mittendorfer, G. S. Hotamisligil, S. Klein, Tauroursodeoxycholic

Acid may improve liver and muscle but not adipose tissue insulin sensitivity in obese men and

women. Diabetes 59, 1899-1905 (2010).

180. A. Crosignani, P. M. Battezzati, K. D. Setchell, P. Invernizzi, G. Covini, M. Zuin, M. Podda,

Tauroursodeoxycholic acid for treatment of primary biliary cirrhosis. A dose-response study.

Dig Dis Sci 41, 809-815 (1996).

181. A. Larghi, A. Crosignani, P. M. Battezzati, G. De Valle, M. Allocca, P. Invernizzi, M. Zuin,

M. Podda, Ursodeoxycholic and tauro-ursodeoxycholic acids for the treatment of primary

biliary cirrhosis: a pilot crossover study. Aliment Pharmacol Ther 11, 409-414 (1997).

Page 115: ATHEROSCLEROSIS IN PROGERIA

VI. REFERENCES

119

182. K. Bentayeb, R. Batlle, C. Sanchez, C. Nerin, C. Domeno, Determination of bile acids in

human serum by on-line restricted access material-ultra high-performance liquid

chromatography-mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 869, 1-

8 (2008).

183. L. R. Hagey, D. L. Crombie, E. Espinosa, M. C. Carey, H. Igimi, A. F. Hofmann,

Ursodeoxycholic acid in the Ursidae: biliary bile acids of bears, pandas, and related

carnivores. Journal of lipid research 34, 1911-1917 (1993).

184. M. Angelico, G. Tisone, L. Baiocchi, G. Palmieri, F. Pisani, S. Negrini, A. Anselmo, G.

Vennarecci, C. U. Casciani, One-year pilot study on tauroursodeoxycholic acid as an adjuvant

treatment after liver transplantation. Italian journal of gastroenterology and hepatology 31,

462-468 (1999).

185. K. D. Setchell, C. M. Rodrigues, M. Podda, A. Crosignani, Metabolism of orally administered

tauroursodeoxycholic acid in patients with primary biliary cirrhosis. Gut 38, 439-446 (1996).

186. P. Invernizzi, K. D. Setchell, A. Crosignani, P. M. Battezzati, A. Larghi, N. C. O'Connell, M.

Podda, Differences in the metabolism and disposition of ursodeoxycholic acid and of its

taurine-conjugated species in patients with primary biliary cirrhosis. Hepatology 29, 320-327

(1999).

187. N. E. Maestri, S. W. Brusilow, D. B. Clissold, S. S. Bassett, Long-term treatment of girls with

ornithine transcarbamylase deficiency. The New England journal of medicine 335, 855-859

(1996).

188. A. B. Burlina, H. Ogier, H. Korall, F. K. Trefz, Long-term treatment with sodium

phenylbutyrate in ornithine transcarbamylase-deficient patients. Molecular genetics and

metabolism 72, 351-355 (2001).

189. K. D. Lindor, E. R. Dickson, W. P. Baldus, R. A. Jorgensen, J. Ludwig, P. A. Murtaugh, J. M.

Harrison, R. H. Wiesner, M. L. Anderson, S. M. Lange, et al., Ursodeoxycholic acid in the

treatment of primary biliary cirrhosis. Gastroenterology 106, 1284-1290 (1994).

190. E. J. Heathcote, K. Cauch-Dudek, V. Walker, R. J. Bailey, L. M. Blendis, C. N. Ghent, P.

Michieletti, G. Y. Minuk, S. C. Pappas, L. J. Scully, et al., The Canadian Multicenter Double-

blind Randomized Controlled Trial of ursodeoxycholic acid in primary biliary cirrhosis.

Hepatology 19, 1149-1156 (1994).

191. R. E. Poupon, A. M. Bonnand, Y. Chretien, R. Poupon, Ten-year survival in ursodeoxycholic

acid-treated patients with primary biliary cirrhosis. The UDCA-PBC Study Group.

Hepatology 29, 1668-1671 (1999).

Page 116: ATHEROSCLEROSIS IN PROGERIA

121

VII. ANNEX

Page 117: ATHEROSCLEROSIS IN PROGERIA

VII. ANNEX

123

Publications related to the thesis:

1. Hamczyk MR, Villa-Bellosta R, Gonzalo P, Andrés-Manzano MJ, López-Otín C, Andrés V.

Ubiquitous and smooth muscle-specific progerin expression in apolipoprotein E-deficient mice

accelerates atherosclerosis and induces premature death (under preparation).

2. Hamczyk MR, Villa-Bellosta R, Quesada V, Gonzalo P, Andrés-Manzano MJ, López-Otín C,

Andrés V. Targeting the ER stress response delays atherosclerosis and associated death in progeroid

mice (under preparation).

Other publications prepared during the thesis:

1. Hamczyk MR*, del Campo L*, Andrés V. Aging in the Cardiovascular System: Lessons from

Hutchinson-Gilford progeria syndrome (under revision in Ann Rev Physiol). * equal contribution

2. Villa-Bellosta R, Hamczyk MR, Andrés V. Novel phosphate-activated macrophages prevent

ectopic calcification by increasing extracellular ATP and pyrophosphate. PLoS One. 2017 Mar

31;12(3):e0174998.

3. Villa-Bellosta R, Hamczyk MR, Andrés V. Alternatively activated macrophages exhibit an

anticalcifying activity dependent on extracellular ATP/pyrophosphate metabolism. Am J Physiol Cell

Physiol. 2016 May 15;310(10):C788-99.

4. Hamczyk MR, Villa-Bellosta R, Andrés V. In Vitro Macrophage Phagocytosis Assay. Methods

Mol Biol. 2015;1339:235-46.

5. Villa-Bellosta R, Hamczyk MR. Isolation and Culture of Aortic Smooth Muscle Cells and In Vitro

Calcification Assay. Methods Mol Biol. 2015;1339:119-29.